US6720306B2 - Tetrapartate prodrugs - Google Patents

Tetrapartate prodrugs Download PDF

Info

Publication number
US6720306B2
US6720306B2 US09/758,993 US75899301A US6720306B2 US 6720306 B2 US6720306 B2 US 6720306B2 US 75899301 A US75899301 A US 75899301A US 6720306 B2 US6720306 B2 US 6720306B2
Authority
US
United States
Prior art keywords
substituted
compound
group
moiety
alkyls
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Expired - Fee Related
Application number
US09/758,993
Other languages
English (en)
Other versions
US20010031873A1 (en
Inventor
Richard B. Greenwald
Hong Zhao
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Enzon Pharmaceuticals Inc
Original Assignee
Enzon Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from US09/183,557 external-priority patent/US6180095B1/en
Application filed by Enzon Pharmaceuticals Inc filed Critical Enzon Pharmaceuticals Inc
Priority to US09/758,993 priority Critical patent/US6720306B2/en
Assigned to ENZON, INC. reassignment ENZON, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: GREENWALD, RICHARD B., ZHAO, HONG
Publication of US20010031873A1 publication Critical patent/US20010031873A1/en
Priority to AU2002239405A priority patent/AU2002239405A1/en
Priority to CA002428018A priority patent/CA2428018A1/en
Priority to EP01987164A priority patent/EP1343494A4/de
Priority to PCT/US2001/045127 priority patent/WO2002043663A2/en
Priority to JP2002545642A priority patent/JP2004518776A/ja
Publication of US6720306B2 publication Critical patent/US6720306B2/en
Application granted granted Critical
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/56Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule
    • A61K47/59Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes
    • A61K47/60Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic macromolecular compound, e.g. an oligomeric, polymeric or dendrimeric molecule obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyureas or polyurethanes the organic macromolecular compound being a polyoxyalkylene oligomer, polymer or dendrimer, e.g. PEG, PPG, PEO or polyglycerol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00

Definitions

  • the present invention relates to tetrapartate prodrugs.
  • the invention relates to polymer conjugates that provide tetrapartate prodrugs that deliver active agents linked to uptake enhancing moieties effective to provide enhanced efficacy, e.g., as antitumor agents or the like.
  • prodrugs include chemical derivatives of a biologically-active material, or parent compound which, upon administration, eventually liberate the parent compound in vivo.
  • Prodrugs allow the artisan to modify the onset and/or duration of action of an agent in vivo and can modify the transportation, distribution or solubility of a drug in the body.
  • prodrug formulations often reduce the toxicity and/or otherwise overcome difficulties encountered when administering pharmaceutical preparations.
  • prodrugs include organic phosphates or esters of alcohols or thioalcohols. See Remington's Pharmaceutical Sciences , 16th Ed., A. Osol, Ed. (1980), the disclosure of which is incorporated by reference herein.
  • Prodrugs are, by definition, forms of the parent or active compound.
  • the rate of release of the active drug is influenced by several factors, but especially by the type of bond joining the active drug to the modifier. Care must be taken to avoid preparing prodrugs which are eliminated through the kidney or reticular endothelial system, etc., before a sufficient amount of the parent compound is released.
  • a polymer as part of the prodrug system, one can increase the circulating half-life of the drug.
  • the double prodrug linkages are selected to hydrolyze in vivo at a rate which generates sufficient amounts of the “second” and more reactive prodrug compound within a suitable time after administration by, e.g., a 1,4-aryl or 1,6-aryl (e.g, benzyl) elimination reaction, providing improved control of the pharmacokinetics of a number of small molecule drugs, agents and the like.
  • a 1,4-aryl or 1,6-aryl (e.g, benzyl) elimination reaction providing improved control of the pharmacokinetics of a number of small molecule drugs, agents and the like.
  • further opportunities for particularly selective targeting of diagnostic and/or therapeutic agents to tissues or cells of interest, by means of a rationally designed prodrug conjugate remain.
  • tumor tissue One particularly desirable target tissue for prodrugs is tumor tissue. It is well known that tumors generally exhibit abnormal vascular permeability characterized by enhanced permeability and retention (“EPR effect”). This EPR effect advantageously allows biologically-effective materials, in the form of macromolecules, e.g., protein(s) such as enzymes and/or antibodies and derivatives or fragments thereof, or the like, to readily enter tumor interstitial tissue space (see, for example, the review article by Maeda et al., 2000 , J. of Controlled Release , 65:271-284, incorporated by reference herein). Certain other tissues, in addition to tumors, can exhibit this same EPR effect, under conditions of inflammation, and the like.
  • EPR effect advantageously allows biologically-effective materials, in the form of macromolecules, e.g., protein(s) such as enzymes and/or antibodies and derivatives or fragments thereof, or the like.
  • the EPR effect allows penetration of large molecule or macromolecule substances, including polymer-based delivery systems. This provides a substantially selective delivery of polymer conjugates into tumor tissue space, e.g., tumor interstitial space. Thereafter, however, the same EPR effect is believed to allow the released prodrug(s) and/or any newly released relatively low molecular weight, biologically-effective materials, to rapidly diffuse out of the extracellular tissue space of the targeted tissue. It is believed that if the released active agent fails to be taken up by the surrounding cells at a sufficient rate, they diffuse away from the release site in the ongoing blood or lymphatic flow.
  • the invention provides for a tetrapartate prodrug in the form of a compound of Formula I:
  • L 1 is a bifunctional linking moiety.
  • D is a moiety that is a leaving group, or a residue of a compound to be delivered into a cell. More particularly, D is a residue of an active biological material, or H and (y) is a positive integer equal to 1 or greater. Preferably, (y) ranges from 1 to about 5. When (y) is greater than 1, each D moiety is independently selected.
  • D can be any biologically active material that it is desired to deliver into a target cell or cells of an animal in need of such treatment, including anti-inflammatory agents, detoxifying agents, anticancer agents, and diagnostics for any of these or other conditions.
  • D is an anticancer agent, an anticancer prodrug, a detectable tag, and combinations thereof.
  • Any anticancer agent or suitable tag that can be linked to the tetrapartate prodrug is contemplated.
  • these include an anthracycline compound, a topoisomerase I inhibitor, daunorubicin, doxorubicin; p-aminoaniline to name but a few.
  • D When D is a leaving group, D can be, e.g., N-hydroxybenzotriazolyl, halogen, N-hydroxyphthalimidyl, p-nitrophenoxy, imidazolyl, N-hydroxysuccinimidyl and/or thiazolidinyl thione.
  • Z is covalently linked to [D] y , wherein Z is a moiety that is actively transported into a target cell, a hydrophobic moiety, and combinations thereof.
  • Z is monovalent, multivalent, or more preferably, bivalent, wherein (y) is 1 or 2.
  • Z itself optionally includes an amino acid residue, a sugar residue, a fatty acid residue, a peptide residue, a C 6-18 alkyl, a substituted aryl, a heteroaryl, —C( ⁇ O), —C( ⁇ S), and —C( ⁇ NR 16 ), where are R 16 is as defined below.
  • the amino acid is, e.g., alanine, valine, leucine, isoleucine, glycine, serine, threonine, methionine, cysteine, phenylalanine, tyrosine, tryptophan, aspartic acid, glutamic acid, lysine, arginine, histidine, proline, and/or a combination thereof, to name but a few.
  • the peptide ranges in size, for instance, from about 2 to about 10 amino acid residues.
  • the peptide is Gly-Phe-Leu-Gly (SEQ ID NO: 1) or Gly-Phe-Leu.
  • Y 1 through Y 4 are independently O, S, or NR 12 ; and R 11 is a mono- or divalent polymer residue.
  • R 1 , R 4 , R 9 , R 10 , R 12 and R 16 are independently hydrogen, C 1-6 alkyl, C 3-12 branched alkyl, C 3-8 cycloalkyl, C 1-6 substituted alkyl, C 3-8 substituted cycloalkyl, aryl, substituted aryl, aralkyl, C 1-6 heteroalkyl, and/or substituted C 1-6 heteroalkyl.
  • R 2 , R 3 , R 5 and R 6 are independently hydrogen, C 1-6 alkyl, C 1-6 alkoxy, phenoxy, C 1-8 heteroalkyl, C 1-8 heteroalkoxy, substituted C 1-6 alkyl, C 3-8 cycloalkyl, C 3-8 substituted cycloalkyl, aryl, substituted aryl, aralkyls, halo-, nitro- and cyano-, carboxy-, C 1-6 carboxyalkyl and/or substituted C 1-6 alkylcarbonyl.
  • Ar is a moiety which, when included in Formula (I), forms a multi-substituted aromatic hydrocarbon or a multi-substituted heterocyclic group; wherein (m), (r), (s), (t), and (u) are independently zero or one, (p) is zero or a positive integer. In certain preferred embodiments, (p) is 1.
  • L 1 is independently one of the following:
  • M is X or Q; where X is an electron withdrawing group;
  • Q is a moiety containing a free electron pair positioned three to six atoms from
  • Q is one of the following: C 2-4 alkyls, cycloalkyls, aryls, and aralkyl groups substituted with a member of the group consisting of NH, O, S, —CH 2 —C(O)—N(H)—, and/or ortho-substituted phenyls;
  • X is, for instance any one of O, NR 20 ,
  • (a) and (n) are independently zero or a positive integer; (b) is zero or one; (g) is a positive integer of 1 or greater;
  • R 7 , R 8 , R 14 , R 15 , R 17 , R 18 and R 20 are independently selected from the same group as that which defines R 1 ; and Y 5 and Y 6 are independently O, S, or NR 12 . It will be appreciated that when (y) is greater than 1, each of the D moieties are the same or different, respectively.
  • (g) ranges from 1 to about 20, or more, but more typically ranges from 1 to about 10.
  • R 11 is a mono- or bivalent polymer, e.g., having a number average molecular weight of from about 2,000 to about 100,000 daltons.
  • the method includes reacting a compound of formula (III):
  • B is a leaving group for Formula (III) and is defined as above for when D is a leaving group.
  • Lx is a leaving group for Formula (IV) and is defined as above for when D is a leaving group.
  • the reaction between (III) and (IV) is preferably conducted in the presence of a solvent and a base.
  • the solvent is, for example, chloroform, methylene chloride, toluene, dimethylformamide and/or combinations thereof Dimethylformamide is generally preferred.
  • the base is, for example, dimethylaminopyridine, diisopropylethylamine, pyridine, triethylamine and/or combinations thereof.
  • Yet another method of preparing a tetrapartate prodrug of the invention is conducted by reacting a compound of formula (V)
  • La is a leaving group as defined when D is a leaving group
  • L 1 , Ar, Z, D, R 1 -R 6 , R 9 -R 11 , Y 1 -Y 3 , and integers are defined as above.
  • the reaction between (V) and the biologically active material is conducted in the presence of a coupling agent, e.g., 1,3-diisopropylcarbodiimide, a dialkyl carbodiimides, 2-halo-1-alkyl-pyridinium halide, 1-(3-dimethylamino-propyl)-3-ethyl carbodiimide, 1-propanephosphonic acid cyclic anhydride, phenyl dichlorophosphates, and/or combinations thereof.
  • a coupling agent e.g., 1,3-diisopropylcarbodiimide, a dialkyl carbodiimides, 2-halo-1-alkyl-pyridinium halide, 1-(3-dimethylamino-propyl)-3-ethyl carbodiimide, 1-propanephosphonic acid cyclic anhydride, phenyl dichlorophosphates, and/or combinations thereof.
  • Methods of using the inventive tetrapartate prodrugs are also provided, e.g., by treating a disease or disorder in an animal, by administering a pharmaceutically acceptable composition comprising an effective amount of a compound of Formula I, to an animal in need thereof.
  • a method is provided of delivering a biologically active material D, into a cell in need of treatment therewith, by a process of:
  • Y* is the remainder of Y 2 , and is independently selected from the group consisting of HO—, HS—, or HNR 12 —;
  • Y** is the remainder of Y*, and is independently selected from the group consisting of O, S, or NR 12 and
  • Z—[D] y crosses the membrane of the cell, and is hydrolyzed therein to release D.
  • FIG. 1 Illustrates a summary of the reaction scheme for the preparation of the tetrapartate prodrugs of doxorubicin by the methods of Examples 1-5, wherein the aromatic is a benzyl derivative.
  • FIG. 2 A Illustrates a general reaction scheme for the degradation of the tetrapartate prodrug prodrug by sequential hydrolysis of the compound of Formula I.
  • variables of FIG. 1 are defined as for Formula I, supra. Symbols for reaction steps: (a): controllable rate in vivo cleavage; (b) “first” prodrug; (c) fast reaction in presence of water; (d) Z—D is the “second” prodrug and it is released substantially into extracellular space; (e) uptake of Z—D into cells and intracellular enzymatic hydrolysis releases D.
  • FIG. 2 B Illustrates a specific reaction schemes for the degradation of tetrapartate prodrug compound identified herein as compounds 14 (top scheme) and 17 (bottom scheme) wherein both schemes result in a final product of doxorubicin substantially released within a cell.
  • Symbols for reaction steps are analogous to those of FIG. 2A, so that (a), (b), (c), (d) and (e) are the analogous reaction steps for the in vivo degradation of compound 14, and that (a′), (b′), (c′), (d′) and (e′) are the analogous reaction steps for the in vivo degradation of compound 17.
  • the products of step (e′) are D and Z, but the figure illustrates that much of Z further degrades into a C 12 acid and leucine.
  • FIG. 3 Illustrates a schematic of the synthesis of compound 2 as described by Method 1 of Example 1.
  • FIG. 4 Illustrates the synthesis of compound 2, as described by Method 2 of Example 1.
  • FIG. 5 Illustrates the synthesis of compound 6, as described by Example 2.
  • FIG. 6 Illustrates a schematic of the synthesis of compound 8, as described by Example 3.
  • FIG. 7 A Illustrates a schematic of the synthesis of compound 10, as described by Example 4.
  • FIG. 7 B Illustrates a schematic of the synthesis of compound 12, as described by Example 5.
  • FIG. 8 Illustrates a schematic of the synthesis of compound 14, as described by Example 6.
  • FIG. 9 Illustrates a schematic of the synthesis of compound 17, as described by Example 7.
  • the invention provides triple prodrugs compositions, hereinafter, “tetrapartate” prodrugs for delivering a biologically active material into certain important target cells, such as, for instance, tumor cells, as well as methods of making and using the same.
  • the tetrapartate prodrug compositions of the present invention contain hydrolyzable linkages between the polymer portion and a biologically active material.
  • the biologically active material is, for example, a moiety derived from a biologically active nucleophile, i.e., a native or unmodified drug or diagnostic tag. These linkages are preferably ester and/or amide linkages designed to hydrolyze at a rate which generates sufficient amounts of the biologically active parent compound in a suitable time.
  • the term “sufficient amounts” for purposes of the present invention shall mean an amount which achieves a therapeutic effect.
  • the present invention is broadly based upon the principle that biologically active materials suitable for incorporation into the polymer-based prodrug conjugates, e.g., the double prodrug compositions as discussed supra, may themselves be substances/compounds which are not active after hydrolytic release from the linked composition, but which will become active after undergoing a further chemical process/reaction, thus providing triple-acting prodrugs.
  • biologically active materials suitable for incorporation into the polymer-based prodrug conjugates e.g., the double prodrug compositions as discussed supra, may themselves be substances/compounds which are not active after hydrolytic release from the linked composition, but which will become active after undergoing a further chemical process/reaction, thus providing triple-acting prodrugs.
  • These triple acting prodrugs are referred to herein as “tetrapartate” prodrugs because the inventive conjugates are provided in essentially four parts.
  • a therapeutic or diagnostic agent that is delivered to the bloodstream by the above-described double prodrug transport system) will remain inactive until entering or being actively transported into a target cell of interest, whereupon it is activated by intracellular chemistry, e.g., by an enzyme or enzyme system present in that tissue or cell.
  • the tetrapartate prodrug conjugates of the invention are thought to provide enhanced effectiveness, e.g., for therapeutic and/or diagnostic activity, in the delivery and activity of certain biologically active materials, e.g., particularly small molecule therapeutic and diagnostic agents.
  • the tetrapartate prodrugs of the invention prepared so that in vivo hydrolysis of the polymer-based conjugate cleaves the conjugate so as to release the active biological material into extracellular fluid, while still linked to the additional moiety.
  • the biologically active materials are preferably, but not exclusively, small molecule therapeutic and/or diagnostic agents. As exemplified below, in one preferred embodiment these are small molecule anticancer agents, and the tissue to be treated is tumor tissue.
  • the rate of transport of a biologically active material into tumor cells is by the delivery of a biologically active material into extracellular tissue space, e.g., of a tissue exhibiting an EPR effect, in a protected and/or transport-enhanced form.
  • extracellular tissue space e.g., of a tissue exhibiting an EPR effect
  • transport enhancers the “additional moiety(s)” as mentioned are described herein as, “transport enhancers.”
  • the transport enhancer is selected from among known substrates for a cell membrane transport system.
  • cells are known to actively transport certain nutrients and endocrine factors, and the like, and such nutrients, or analogs thereof, are readily employed to enhance active transport of a biologically effective material into target cells.
  • these nutrients include amino acid residues, peptides, e.g., short peptides ranging in size from about 2 to about 10 residues or more, simple sugars and fatty acids, endocrine factors, and the like.
  • Desirable amino acid residues include all of the known naturally-occurring L-amino acids.
  • L-isoleucine as a transport enhancer is exemplified in the Examples provided below.
  • D-amino acids are useful as transport enhancers, e.g., both D and L-alanine, and other analogous amino acid optical isomers, show the same activity.
  • Derivatives and analogs of the naturally occurring amino acids, as well as various art-known non-naturally occurring amino acids (D or L), hydrophobic or non-hydrophobic are also contemplated to be within the scope of the invention.
  • amino acid analogs and derivates include: 2-aminoadipic acid, 3-aminoadipic acid, beta-alanine, beta-aminopropionic acid, 2-aminobutyric acid, 4-aminobutyric acid, piperidinic acid, 6-aminocaproic acid, 2-aminoheptanoic acid, 2-aminoisobutyric acid, 3-aminoisobutyric acid, 2-aminopimelic acid, 2,4-diaminobutyric acid, desmosine, 2,2-diaminopimelic acid, 2,3-diaminopropionic acid, n-ethylglycine, N-ethylasparagine, 3-hydroxyproline, 4-hydroxyproline, isodesmosine, allo-isoleucine, N-methylglycine, sarcosine, N-methylisoleucine, 6-N-methyllysine, N-methylvaline, norvaline, norleucine, orn
  • Short peptides are, for example, peptides ranging from 2 to about 10, or more, amino acid residues, as mentioned supra.
  • peptide transport enhancers need not be hydrophobic, but are thought to function in other ways to enhance uptake and/or to protect the linked small molecule agents from premature hydrolysis in the general bloodstream.
  • peptide transport enhancers, and other transport enhancers of similar molecular weight ranges are thought to sterically hinder cleavage from the biologically active agent by plasma-based hydrolytic enzymes, but are then cleaved within a target cell by various peptides and/or proteases, such as cathepsins.
  • the transport enhancer is a hydrophobic moiety.
  • a hydrophobic moiety inhibits the extracellular cleavage of the transport enhancer away from the active biological agent, by inhibiting the attack of hydrolytic enzymes, etc. present in the extracellular tissue space, e.g., in the plasma.
  • preferred transport enhancers include, e.g., hydrophobic amino acids such as alanine, valine, leucine, isoleucine, methionine, proline, phenylalanine, tyrosine, and tryptophane, as well as non-naturally occurring derivatives and analogs thereof, as mentioned supra.
  • the transport enhancer is a hydrophobic organic moiety.
  • the organic moiety is a C 6-18 , or larger, alkyl, aryl or heteroaryl-substituted or nonsubstituted.
  • the organic moiety transport enhancer is also contemplated to encompass and include organic functional groups including,
  • tetrapartate refers to prodrug conjugates, and in particular, to conjugates incorporating the features of the double prodrugs, as discussed supra, and an additional moiety serving as a transport enhancer positioned between the residue of the biologically active compound and the polymer moiety to form a 4-part structure wherein the biologically active agent is the fourth part of the conjugate.
  • This structure provides that the residue of the biologically active compound is optimized for transport and release substantially into a target cell.
  • the fourth element of the “tetrapartate” is therefore the residue of the biologically active compound, itself.
  • diagnostic tetrapartate conjugates incorporating detectable tags are also contemplated, and the use of the terms, “tetrapartate prodrug,” or simply, “prodrug” herein, with reference to the inventive conjugates, broadly also includes conjugates and methods of making and delivering diagnostic reagents, including tagged drugs, as well, unless otherwise specified or distinguished.
  • biologically active material and “biologically active compound,” and/or “biologically active agent,” etc., are used interchangeably unless otherwise stated. These terms refer, for example, to a drug or pharmaceutical, and/or a diagnostic agent or reagent, such as a detectable label or marker.
  • diagnostic agent or reagent such as a detectable label or marker.
  • active agent refers to compound(s) with useful activity, particularly when administered to an animal, in vivo, and/or to precursors of the same, unless otherwise stated.
  • biological activity is any property of such a material or compound that is useful in an animal or person, e.g., for medical, and/or diagnostic purposes.
  • the biological activity is manifested in the intracellular space, i.e., the drug or diagnostic agent preferably but not exclusively is useful once delivered/released into the cytoplasm and/or nucleus of one or more types of target cell of interest.
  • the use of the singular or plural is not meant to be limiting of the numerical number of the referenced item or object.
  • the use of the singular to refer to a cell, polymer or drug does not imply that only one cell is treated, only one molecule is prepared or employed, and/or only one drug is employed, and the use of the plural does not exclude application to a single referenced item, unless expressly stated.
  • the terms, “cell,” “cell type,” “target cell,” and etc. are used interchangeably unless otherwise specified and refer to both singular and plural cells, however organized into a tissue, tissues or other system or component, normal or pathological, of an animal or patient to be treated.
  • the term “residue” shall be understood to mean that portion of a biologically active compound which remains after it has undergone a reaction in which the prodrug carrier portion has been attached by modification of e.g., an available hydroxyl or amino group, to form, for example, an ester or amide group, respectively.
  • alkyl shall be understood to include, e.g., straight, branched, substituted C 1-12 alkyls, including alkoxy, C 3-8 cycloalkyls or substituted cycloalkyls, etc.
  • the prodrugs of the present invention include the double prodrugs taught by co-owned Ser. Nos. 09/183,557 now U.S Pat. No. 6,180,095 and Ser. No. 08/992,435, now abandoned it is generally preferred that the polymeric portion is first released by hydrolysis and then the resultant “second prodrug” moiety undergoes a 1,4- or 1,6-aryl (e.g., benzyl) elimination reaction to regenerate, for example, a moiety comprising a further prodrug, Thereafter, the released moiety diffuses and/or is transported into target cells, where a substantial proportion of the incorporated remainder of the prodrug is further cleaved or hydrolyzed by intracellular enzymes to release the biologically active compound.
  • a 1,4- or 1,6-aryl e.g., benzyl
  • cancer or “tumor” are clinically descriptive terms which encompass a myriad of diseases characterized by cells that exhibit unchecked and abnormal cellular proliferation.
  • tissue when applied to tissue, generally refers to any abnormal tissue growth, i.e., excessive and abnormal cellular proliferation.
  • cancer is an older term which is generally used to describe a malignant tumor or the disease state arising therefrom.
  • the art refers to an abnormal growth as a neoplasm, and to a malignant abnormal growth as a malignant neoplasm.
  • the compounds according to the invention are as follows.
  • L 1 is a bifunctional linking moiety; for example, L 1 is independently one of the following:
  • L 1 is
  • Z is a transport enhancer and/or protector that is covalently linked to D, wherein Z is selected to enhance or improve intracellular delivery of Z—[D] y into a cell; relative to the intracellular delivery of D without Z.
  • Z optionally includes one of the following: an amino acid residue, a sugar residue, a fatty acid residue, a peptide residue, a C 6-18 alkyl, a substituted aryl, a heteroaryl, —C( ⁇ O), —C( ⁇ S), and —C( ⁇ NR 16 ), and/or combinations thereof.
  • D is a moiety that is a leaving group, or a residue of a compound to be delivered into a cell.
  • D is an active biological material, or H.
  • each D can be selected independently, so that there can be as many as five, or more, different types of moieties linked to Z for delivery into a target cell of interest.
  • D is a therapeutic agent or drug, but D is also optionally a diagnostic agent.
  • D can be two moieties, including both a therapeutic agent and a diagnostic tag for delivery into the same cell type or into the cells of a tissue type of interest. Further still, such plural D moieties will comprise multiple different therapeutic agents, preferably targeted to a single type of cell, where when delivered and released together, the different agents act synergistically to achieve a desired therapeutic effect.
  • D is one or more anticancer agent(s) and/or an anticancer prodrug, or residue thereof.
  • D is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl
  • B is H, a leaving group, a residue of an amine-containing moiety, or a residue of a hydroxyl-containing moiety;
  • Y 1-6 are independently O, S or NR 12 ;
  • M is X or Q
  • X is an electron withdrawing group
  • Q is a moiety containing a free electron pair positioned three to six atoms from
  • R 1 , R 4 , R 7 , R 8 , R 9 , R 10 , R 12 , R 14 , R 15 , R 16 and R, 18 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 3-12 branched alkyls, C 3-8 cycloalkyls, C 1-6 substituted alkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, C 1-6 heteroalkyls and substituted C 1-6 heteroalkyls;
  • R 2 , R 3 , R 5 and R 6 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 1-6 alkoxys, phenoxy, C 1-8 heteroalkyls, C 1-8 heteroalkoxy, substituted C 1-6 alkyls, C 3-8 cycloalkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, halo-, nitro- and cyano-, carboxy-, C 1-6 carboxyalkyls and C 1-6 alkylcarbonyls;
  • Ar is a moiety which when included in Formula (I) forms a multi-substituted aromatic hydrocarbon or a multi-substituted heterocyclic group;
  • (p) is zero or a positive integer
  • R 11 is a polymer such as a polyalkylene oxide.
  • the Ar is a moiety, which when included in Formula (I), forms a multi-substituted aromatic hydrocarbon or a multi-substituted heterocyclic group.
  • a key feature is that the Ar moiety is aromatic in nature. Generally, to be aromatic, the ⁇ -electrons must be shared within a “cloud” both above and below the plane of a cyclic molecule. Furthermore, the number of ⁇ electrons must satisfy the Huckle rule (4n+2). Those of ordinary skill will realize that a myriad of moieties will satisfy the aromatic requirement of the moiety and thus are suitable for use herein.
  • Preferred aromatic hydrocarbon moieties include, without limitation:
  • J is O, S, or N—R 19
  • E and Z are independently C—R 19 or N—R 19
  • R 19 is independently selected from the same group as that which defines R 9 in Formula (I) e.g., hydrogen, C 1-6 alkyls, etc.
  • Isomers of the five and six-membered rings are also contemplated as well as benzo- and dibenzo- systems and their related congeners are also contemplated. It will also be appreciated by the artisan of ordinary skill that the aromatic rings can optionally be substituted with hetero-atoms such as O, S, NR 13 , etc. so long as Huckel's rule is obeyed.
  • aromatic or heterocyclic structures may optionally be substituted with halogen(s) and/or side chains as those terms are commonly understood in the art.
  • all structures suitable for Ar moieties of the present invention are capable of allowing the Y 3 and C(R 1 )(R 4 ) moieties to be in a para or an ortho arrangement with the same plane as shown in Formulas I-A and I-B, below.
  • preferred aspects of the present invention define (r), (s), (t), and (u) as one and R 2 and R 6 as being independently selected from the group consisting of methyl, C 1-6 alkyls, methyl, C 1-6 alkoxys, and methoxy. More preferably, R 2 and R 6 are either both methyl or methoxy moieties.
  • R 3 and R 5 are preferably both hydrogen
  • R 1 and R 4 are preferably either hydrogen, CH 3 or CH 2 CH 3 .
  • Y 1 through Y 4 i.e., Y 1-4
  • substituted alkyls include carboxyalkyls, aminoalkyls, dialkylaminos, hydroxyalkyls and mercaptoalkyls; substituted cycloalkyls include moieties such as 4-chlorocyclohexyl; aryls include moieties such as napthyl; substituted aryls include moieties such as 3-bromophenyl; aralkyls include moieties such as toluyl; heteroalkyls include moieties such as ethylthiophene; substituted heteroalkyls include moieties such as 3-methoxy-thiophene; alkoxy includes moieties such as methoxy; and phenoxy includes moieties such as 3-nitrophenoxy.
  • Halo- shall be understood to include fluoro, chloro, iodo and bromo.
  • the invention includes bifunctional linking moiety L 1 which when combined with
  • Suitable amino acid residues can be selected from naturally-occurring or synthetic, i.e. non-naturally-occurring, amino acids including alanine, valine, leucine, isoleucine, glycine, serine, threonine, methionine, cysteine, phenylalanine, tyrosine, tryptophan, aspartic acid, glutamic acid, lysine, arginine, histidine or proline.
  • Some preferred peptide residues include Gly-Phe-Leu-Gly (SEQ ID NO: 1) and Gly-Phe-Leu. It is noted that the terminal amino group of the amino acid or peptide residue will be proximal to R 11 (i.e. polymer).
  • Peptides can be readily synthesized or obtained from commercial sources for inclusion herein.
  • L 1 includes the moiety (M) which is either an electron withdrawing group (designated herein as X), or a moiety containing a free electron pair positioned three to six atoms from the
  • the tetrapartate conjugates of the invention are based upon an aromatic moiety that is a substituted benzyl, and the first two breaks in the prodrug, in vivo, are based on a 1,4 or 1,6 benzyl elimination mechanism.
  • This embodiment provides a conjugate as illustrated by FIG. 1 .
  • FIG. 1 an overview of the general reaction scheme followed in synthesis of the compounds produced in Examples 1-5, provided herein below, is set forth.
  • Precursor compound (FIG. 1, compound a) is reacted in the presence of leucine doxorubicin (“leu-dox”), dimethylaminopyridine and dimethyformamide to form (FIG. 1, compound b), wherein the variables are set forth by Table 1, below.
  • (n) is an integer from 1 to about 12. More preferably, (n) is 1 or 2.
  • the moiety may be an electron withdrawing group, designated herein as X.
  • electron withdrawing groups are groups which tend to pull shared electrons toward themselves thereby making carbon more electro-positive. This, in turn, destabilizes the carbonyl moiety, causing more rapid hydrolysis.
  • X when X is in the ⁇ position to the ester, it modulates the rates of hydrolysis and enzymatic cleavage.
  • X can be moieties such as O, NR 20 ,
  • R 6 is the same as that defined by Y 1 , R 12 and R 17 are the same as defined above i.e., H, C 1-6 alkyls, branched alkyls, aryls, etc.
  • R 1 is the same as defined by Formula I, supra.
  • R 20 is H, a C 1-6 alkyl such as methyl or ethyl or substituted C 1-6 alkyl. It is preferred that X is either O or NR 20 .
  • L 1 includes Q, which is a moiety containing a free electron pair positioned three to six atoms from the
  • the polymer, R 11 is preferably attached to Q via a heteroatom such as oxygen.
  • the free electron pair is five atoms from this oxygen.
  • Q can be selected from the non-limiting list of C 2-4 alkyls or cycloalkyls, aryls or aralkyl groups substituted with a member of the group consisting of O, S and NR 12 .
  • the free electron pair can be anywhere along the Q moiety as long as the defined spacing between the free electron pair and Y 4 is maintained.
  • R 11 is attached to Q via NR 12 , O, or S.
  • Q assists hydrolysis of the prodrug linkage by anchimeric assistance because the free electron pair moiety can generate a three- to six-membered, but preferably five-membered, ring by-product upon hydrolysis of the preferably ester linkage.
  • Q can also be selected from the group consisting of C 2-4 alkyls, cycloalkyls, aryls, aralkyl groups substituted with a member of the group consisting of NH, O, S, —CH 2 —C(O)—N(H)—, and ortho-substituted phenyls such as
  • R 21 is selected from the same group as that which defines R 12 supra and T is any moiety linked to Q.
  • the prodrug compounds of the present invention are designed so that the t 1/2 of hydrolysis is less than the t 1/2 of elimination in plasma.
  • the linkages included in the compounds have in-vivo hydrolysis rates, in plasma, that are short enough to allow sufficient amounts of the transport enhanced conjugate with parent compounds, i.e., the amino- or hydroxyl-containing biologically active compound, to be released prior to elimination.
  • Some preferred compounds of the present invention i.e., those in which (n) is 1, have a t 1/2 for hydrolysis in plasma ranging from about 5 minutes to about 12 hours.
  • the compositions have a plasma t 1/2 of hydrolysis ranging from about 0.5 to about 8 hours and most preferably from about 1 to about 6 hours.
  • the polymeric residue is cleaved and the resultant second prodrug moiety remains.
  • various intracellular peptidases and/or proteases including e.g., cathepsins, cleave, e.g., by enzymatic hydrolysis, the transport enhancer moiety to release the biologically effective material within the target cell.
  • the following degradation scheme is provided for illustrative purposes and is not intended to limit the scope of the invention.
  • FIG. 2A illustrates, in overview, an in vivo degradation reaction is shown wherein the variables of the starting tetrapartate compound are defined as described supra for Formula I.
  • the illustrated tetrapartate prodrug undergoes a controllable rate cleavage, in vivo, labeled as (a) to remove R 11 .
  • the remaining compound (b) immediately undergoes a fast hydrolysis (c) in the presence of water to separate (b) and release Z—D, the enhancer-prodrug is released (d) mostly into the extracellular tissue space as Z—D.
  • Z—D is, in turn, taken up by surrounding cells and hydrolyzed intracellularly (e) to release D. Any D that might be released in the intracellular tissue space is thought to be rapidly removed by blood or plasma flow, and is believed to provide little or no added benefit in terms of a therapeutic or diagnostic function.
  • FIG. 2B provides schematics of in vivo degradation reactions thought to occur for compounds 14 and 17 (see, e.g., the Examples below for synthetic details).
  • compound 14 is degraded by a controllable rate cleavage, in vivo, labeled as (a) to remove PEG.
  • the remaining compound (b) immediately undergoes a fast hydrolysis (c) in the presence of water to separate (b) and release inter alia, (d) which is analogous to Z—D of FIG. 2 A.
  • the enhancer-doxorubicin prodrug is released mostly into the extracellular tissue space (d).
  • the enhancer-doxorubicin prodrug is, in turn, taken up by surrounding cells and hydrolyzed intracellularly (e) to release active doxorubicin.
  • An analogous degradation process is described in FIG. 2B for compound 17, wherein the analogous process steps are labeled as (a′), (b′), (c′), (d′) and (e′).
  • Reaction (f) and (f′) refers to respective side reactions wherein the aromatic remainder that is cleaved from the prodrug is converted to a water soluble hydroxyl derivative.
  • the “tetrapartate prodrug” compositions of the present invention include water-soluble polymer, R 11 .
  • R 11 includes a capping group A.
  • Capping group A includes, for example, hydrogen, C 1-6 alkyl moieties, carboxyalkyl, dialkyl acyl urea alkyls, and/or a compound of formula (II) shown below, which forms a bis-system:
  • G′ is the same as D or another member of the group defined by D, v is 0 or 1, and the remaining variables are as set forth above with regard to Formula (I).
  • Suitable examples of such polymers include polyalkylene oxides such as polyethylene glycols which are also preferably substantially non-antigenic.
  • the general formula for PEG and its derivatives i.e. A′—O—(CH 2 CH 2 O) x —(CH 2 ) n —A, where (x) represents the degree of polymerization (i.e. 10-2,300) or number of repeating units in the polymer chain and is dependent on the molecular weight of the polymer; (n) is zero or a positive integer; A is a capping group as defined herein, i.e. an —H, amino, carboxy, halo, C 1-6 alkyl or other activating group and A′ is the same as A or another A moiety.
  • polypropylene glycols branched PEG derivatives such as those described in commonly-assigned U.S. Pat. No. 5,643,575, “star-PEG's” and multi-armed PEG's such as those described in Shearwater Polymers, Inc. catalog “Polyethylene Glycol Derivatives 1997-1998”; the disclosure of each is incorporated herein by reference.
  • the water-soluble polymer will be functionalized for attachment to the linkage via M, X or Q herein.
  • the PEG portion of the prodrugs can be the following non-limiting compounds:
  • Y 7 is O or S; and A, R 12 , (n) and (x) are as defined above.
  • polyethylene glycols PEG's
  • mono-activated, C 1-4 alkyl-terminated PAO's such as mono-methyl-terminated polyethylene glycols (mPEG's) are preferred when mono-substituted polymers are desired; bis-activated polyethylene oxides are preferred when di-substituted prodrugs are desired.
  • mono- or di-acid activated polymers such as PEG acids or PEG diacids can be used as well as mono-or di-PEG amines and mono- or di-PEG diols.
  • Suitable PAO acids can be synthesized by first converting mPEG—OH to an ethyl ester followed by saponification. See also Gehrhardt, H., et al. Polymer Bulletin 18: 487 (1987) and Veronese, F. M., et al., J. Controlled Release 10; 145 (1989).
  • the PAO-acid can be synthesized by converting mPEG—OH into a t-butyl ester followed by acid cleavage. See, for example, commonly assigned U.S. Pat. No. 5,605,976. The disclosures of each of the foregoing are incorporated by reference herein.
  • PAO's and PEG's can vary substantially in number average molecular weight, polymers ranging from about 2,000 to about 100,000 daltons are usually selected for the purposes of the present invention. Molecular weights of from about 20,000 to about 50,000 are preferred, and 20,000 to about 40,000 are particularly preferred. The number average molecular weight of the polymer selected for inclusion in the “tetrapartate prodrug” must be sufficient so as to provide sufficient circulation of the “tetrapartate prodrug” before removal of the transport enhancer. Within the ranges provided above, polymers having molecular weight ranges of at least 20,000 are preferred in some aspects for chemotherapeutic and organic moieties. In the case of some nucleophiles such as certain proteins, enzymes and the like, polymers having a molecular weight range of from about 12,000 to about 20,000 are preferred.
  • the polymeric substances included herein are preferably water-soluble at room temperature.
  • a non-limiting list of such polymers include polyalkylene oxide homopolymers such as polyethylene glycol (PEG) or polypropylene glycols, polyoxyethylenated polyols, copolymers thereof and block copolymers thereof, provided that the water solubility of the block copolymers is maintained.
  • effectively non-antigenic materials such as dextran, polyvinyl alcohols, carbohydrate-based polymers, hydroxypropylmethacrylamide (HPMA), and copolymers thereof, etc. and the like can be used if the same type of activation is employed as described herein for PAO's such as PEG.
  • PAO's such as PEG.
  • one method includes reacting a compound of formula (III).
  • L 1 is a bifunctional linking moiety:
  • B and Lx are independently selected leaving groups and are defined as above for when D is a leaving group
  • Z is a moiety that is actively transported into a target cell, a hydrophobic moiety and combinations thereof.
  • Z is selected from the group consisting of an amino acid residue, a C 6-18 alkyl, a substituted aryl, a hetero aryl, —C( ⁇ Y 4 ), —C( ⁇ S), —C( ⁇ NR 16 ) and combinations thereof, wherein R 16 is selected from the same group as R 12 ;
  • R 1 , R 4 , R 9 , R 10 and R 12 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 3-12 branched alkyls, C 3-8 cycloalkyls, C 1-6 substituted alkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, C 1-6 heteroalkyls and substituted C 1-6 heteroalkyls;
  • R 2 , R 3 , R 5 and R 6 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 1-6 alkoxys, phenoxy, C 1-8 heteroalkyls, C 1-8 heteroalkoxys, substituted C 1-6 alkyls, C 3-8 cycloalkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, halo-, nitro- and cyano-, carboxy-, C 1-6 carboxyalkyls and C 1-6 alkylcarbonyls;
  • Ar is a moiety which when included in Formula (III), and subsequently in Formula (I), forms a multi-substituted aromatic hydrocarbon or a multi-substituted heterocyclic group;
  • (p) is zero or a positive integer
  • Y 1-4 are independently O, S, or NR 12 , wherein the definition of R 12 is defined as per formula I, supra; (y) is one or two; and R 11 is a monovalent or divalent polymer residue.
  • the reaction between (III) and (IV) is conducted in the presence of a solvent and a base.
  • the solvent is preferably an inert solvent, i.e., inert with respect to the reactants and products.
  • exemplary solvents include, simply by way of example, chloroform, toluene, methylene chloride, dimethylformamide and combinations thereof. Generally, dimethylformamide is preferred.
  • Exemplary bases solvents include, simply by way of example, dimethylaminopyridine, diisopropylethylamine, pyridine, triethylamine and combinations thereof Generally, dimethylaminopyridine is preferred.
  • the tetrapartate prodrugs of the invention can also be prepared by reacting a compound of formula (V)
  • a biologically effective material e.g., a biologically active compound such as a drug or diagnostic tag.
  • a biologically effective material e.g., a biologically active compound such as a drug or diagnostic tag.
  • La is a leaving group for Formula V as defined for D when D is a leaving group.
  • L 1 is a bifunctional linking moiety
  • Z is a moiety that is actively transported into a target cell, a hydrophobic moiety and combinations thereof
  • Z is selected from the group consisting of an amino acid residue, a C 6-18 alkyl, a substituted aryl, a hetero aryl, —C( ⁇ Y 4 ), —C( ⁇ S), —C( ⁇ NR 16 ) and combinations thereof;
  • R 1 , R 4 , R 9 , R 10 , R 12 and R 16 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 3-12 branched alkyls, C 3-8 cycloalkyls, C 1-6 substituted alkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, C 1-6 heteroalkyls and substituted C 1-6 heteroalkyls;
  • R 2 , R 3 , R 5 and R 6 are independently selected from the group consisting of hydrogen, C 1-6 alkyls, C 1-6 alkoxys, phenoxy, C 1-8 heteroalkyls, C 1-8 heteroalkoxys, substituted C 1-6 alkyls, C 3-8 cycloalkyls, C 3-8 substituted cycloalkyls, aryls, substituted aryls, aralkyls, halo-, nitro- and cyano-, carboxy-, C 1-6 carboxyalkyls and C 1-6 alkylcarbonyls.
  • Ar is a moiety which when included in Formula (V) forms a multi-substituted aromatic hydrocarbon or a multi-substituted heterocyclic group.
  • (p) is zero or a positive integer.
  • Y 1-4 are independently O, S, or NR 12 , wherein the definition of R 12 is defined as per formula I, supra; and R 11 is a monovalent or divalent polymer residue.
  • the reaction between (V) and the biologically effective material is conducted in the presence of a coupling agent, including, for example, 1,3-diiso-propylcarbodiimide, a dialkyl carbodiimides, 2-halo-1-alkyl-pyridinium halide, 1-(3-dimethylamino-propyl)-3-ethyl carbodiimide, 1-propanephosphonic acid cyclic anhydride, phenyl dichlorophosphates, and combinations thereof.
  • the reaction between (V) and the biologically active material is typically conducted in the presence of a solvent and a base, each of which are defined as described supra, for the reaction between formulas (III) and (IV).
  • the base is preferably dimethylamino pyridine.
  • suitable leaving groups include, without limitations, moieties such as N-hydroxybenzotriazolyl, halogen, N-hydroxyphthalimidyl, p-nitrophenoxy, imidazolyl, N-hydroxysuccinimidyl; thiazolidinyl thione, or other good leaving groups as will be apparent to those of ordinary skill.
  • moieties such as N-hydroxybenzotriazolyl, halogen, N-hydroxyphthalimidyl, p-nitrophenoxy, imidazolyl, N-hydroxysuccinimidyl; thiazolidinyl thione, or other good leaving groups as will be apparent to those of ordinary skill.
  • the synthesis reactions used and described herein will be understood by those of ordinary skill without undue experimentation.
  • the acylated intermediate compound (III) can be reacted with such as 4-nitrophenyl chloroformate, disuccinimidyl carbonate (DSC), carbonyldiimidazole, thiazolidine thione, etc. to provide the desired activated derivative.
  • DSC disuccinimidyl carbonate
  • carbonyldiimidazole thiazolidine thione, etc.
  • acylation of the p-hydroxybenzyl alcohol or the p-aminobenzyl alcohol and the o-hydroxbenzyl alcohol or the o-aminobenzyl alcohol can be carried out with, for example, thiazolidine thione activated polymers, succinimidyl carbonate activated polymers, carboxylic acid activated polymers, blocked amino acid derivatives.
  • the “activated” form of the PEG prodrug (or blocked prodrug) is ready for conjugation with an amine- or hydroxyl-containing compound.
  • Some preferred activated transport forms are shown below.
  • the only limitations on the types of biologically effective materials suitable for inclusion herein is that there is available at least one site for covalent attachment to the uptake enhancer moiety.
  • this can be a (primary or secondary) amine-containing position or functional group which can react and link with a carrier portion, e.g., by forming an amide bond.
  • Other sites for covalent attachment to the uptake enhancer moiety include, e.g., a hydroxyl functional group, to form, e.g., an ester linkage.
  • the selected linkage between the biologically effective material of interest, and the uptake enhancer is such that there is no substantial loss of bioactivity after the double prodrug portion of the conjugate releases the parent compound in linkage with a transport enhancer.
  • the remaining amine-containing compound is referred to as the residue of the unconjugated compound.
  • D is a residue of an amine-containing organic compound.
  • Organic compounds include, without limitation, moieties such as anthracycline compounds including daunorubicin, doxorubicin; p-aminoaniline mustard, melphalan, Ara-C (cytosine arabinoside) and related anti-metabolite compounds, e.g., gemcitabine, etc.
  • biologically effective material is a biologically active compound that is suitable for medicinal or diagnostic use in the treatment of animals, e.g., avians and/or mammals, including humans, for conditions for which such treatment is desired.
  • animals e.g., avians and/or mammals, including humans
  • biologically active materials are meant to be illustrative and not limiting for the compounds which can be modified. Those of ordinary skill will realize that other such compounds can be similarly modified without undue experimentation. It is to be understood that those biologically active materials not specifically mentioned but having suitable amino-groups are also intended and are within the scope of the present invention.
  • Camptothecin is a water-insoluble cytotoxic alkaloid produced by Camptotheca accuminata trees indigenous to China and nothapodytes foetida trees indigenous to India. Camptothecin and related compounds and analogs are also known to be potential anticancer or antitumor agents and have been shown to exhibit these activities in vitro and in vivo. Camptothecin and related compounds are also candidates for conversion to the tetrapartate prodrugs of the present invention.
  • Camptothecin and certain related analogues share the structure:
  • the A ring in either or both of the 10- and 11-positions can be substituted with an OH.
  • the A ring can also be substituted in the 9-position with a straight or branched C 1-30 alkyl or C 1-17 alkoxy, optionally linked to the ring by a heteroatom i.e. —O or S.
  • the B ring can be substituted in the 7-position with a straight or branched C 1-30 alkyl or substituted alkyl-, C 5-8 cycloalkyl, C 1-30 alkoxy, phenyl alkyl, etc., alkyl carbamate, alkyl carbazides, phenyl hydrazine derivatives, amino-, aminoalkyl-, aralkyl, etc. Other substitutions are possible in the C, D and E rings.
  • camptothecin derivatives for use herein include those which include a 20-OH or another OH moiety which is capable of reacting directly with activated forms of the polymer transport systems described herein or to the linking moiety intermediates, e.g. iminodiacetic acid, etc., which are then attached to a polymer such as PEG.
  • camptothecin analogs herein has been made for purposes of illustration and not limitation.
  • taxanes One class of compounds included in the tetrapartate prodrug compositions of the present invention is taxanes.
  • taxanes include all compounds within the taxane family of terpenes.
  • taxol paclitaxel
  • 3′-substituted tert-butoxy-carbonyl-amine derivatives taxoteres
  • other analogs which are readily synthesized using standard organic techniques or are available
  • Paclitaxel R′ 1 ⁇ C 6 H 5 ; R′ 2 ⁇ CH 3 CO; Taxotere: R′ 1 ⁇ (CH 3 ) 3 CO; R′ 2 ⁇ H
  • the tetrapartate prodrug formulations of the present invention can be prepared using many other compounds.
  • drugs such as gemcitabine, etoposide, triazole-based antifungal agents such as fluconazole and/or ciclopirox can be used.
  • the parent compounds selected for tetrapartate prodrug forms need not be substantially water-insoluble, although the polymer-based tetrapartate prodrugs of the present invention are especially well suited for delivering such water-insoluble compounds.
  • Other useful parent compounds include, for example, certain low molecular weight biologically active proteins, enzymes and peptides, including peptido glycans, as well as other anti-tumor agents; cardiovascular agents such as forskolin; anti-neoplastics such as combretastatin, vinblastine, doxorubicin, ara-C, maytansine, etc.; anti-infectives such as vancomycin, erythromycin, etc.; anti-fungals such as nystatin, amphoteracin B, triazoles, papulocandins, pneumocandins, echinocandins, polyoxins, nikkomycins, pradimicins, benanomicins, etc.
  • anti-anxiety agents include gastrointestinal agents, central nervous system-activating agents, analgesics, fertility or contraceptive agents, anti-inflammatory agents, steroidal agents, anti-urecemic agents, cardiovascular agents, vasodilating agents, vasoconstricting agents and the like.
  • the remaining amine-or hydroxyl-containing compound is referred to as the residue of the unconjugated compound.
  • hybrid types of the polymeric tetrapartate prodrug transport system described herein include not only the reversible double prodrug system described above but also a second polymeric transport system based on more permanent types of linkages.
  • the hybrids can be prepared by at least two methods.
  • the benzyl-elimination-based prodrug can be synthesized first and then PEGylated using any art-recognized activated polymer such as thiazolidinyl thione-or succinimidyl carbonate-activated PEG.
  • the more permanent conjugation reaction can be performed first and the resultant conjugates can be used to form the double prodrug portion of the tetrapartate conjugates described herein.
  • the hybrid systems will be better suited for proteins, enzymes and the like where multiple amino groups are available for attachment of the polymeric transport forms.
  • “activated polymers” will be understood to include polymers containing one or more terminal groups which are capable of reacting with one or more of ⁇ -amino groups, ⁇ -amino groups, histidine nitrogens, carboxyl groups, sulfhydryl groups, etc. found on enzymes, proteins, etc., as well as such groups found on synthetically prepared organic compounds. It will further be appreciated that the activating groups described below can also be used to form the activated transport forms described above.
  • the activating terminal moiety can be any group which facilitates conjugation of the polymers with the biologically active material, i.e., protein, enzyme, etc. either before of after the double prodrug transport system of the present invention has been synthesized. See, for example, U.S. Pat. No. 4,179,337, the disclosure of which is hereby incorporated by reference.
  • Such activating groups can be a moiety selected from:
  • active esters such as N-hydroxy-succinimidyl or N-hydroxybenzotriazolyl.
  • hydrazine and hydrazide functional groups such as the acyl hydrazides, carbazates, semicarbamates, thiocarbazates, etc.
  • Functional groups capable of reacting with hydroxyl groups such as (carboxylic) acids or other nucleophiles capable of reacting with an electrophilic center.
  • a non-limiting list includes, for example, hydroxyl, amino, carboxyl, thiol groups, active methylene and the like.
  • the activating moiety can also include a spacer moiety located proximal to the polymer.
  • the spacer moiety may be a heteroalkyl, alkoxy, alkyl containing up to 18 carbon atoms or even an additional polymer chain.
  • the spacer moieties can added using standard synthesis techniques.
  • another aspect of the present invention provides methods for delivering biologically active materials, such as therapeutic or diagnostic agents into cells where such biological activity is desired.
  • biologically active materials such as therapeutic or diagnostic agents
  • the tetrapartate prodrugs of the invention are readily employed to deliver biologically active materials into a wide variety of cells, found throughout the animal body, certain applications are preferred.
  • the tetrapartate prodrugs of the invention are particularly useful in delivering biologically active materials, such as drugs and/or diagnostics, into cells present in tissues exhibiting the above-discussed EPR effect.
  • tissue types exhibiting EPR occur in different diseases and disorders, including tissues undergoing inflammation, toxic reactions of various kinds, as well as solid tumors.
  • the broad method includes contacting living tissue with the inventive tetrapartate prodrugs.
  • tissue Preferably the tissue exhibits the EPR effect, so that polymer linked conjugates preferably enter such tissues.
  • an agent once delivered into a target cell and activated, can then be released by that cell and provide biological activity in other tissue spaces.
  • a non-active prodrug of the invention that is delivered into an exocrine cell of the liver or pancreas under suitable conditions, e.g., during a disease process that causes inflammation, and results in an EPR effect, can be activated within the cytoplasm of the target cell, and then the activated drug or diagnostic agent can then be secreted into the gastrointestinal (“G.I”) tract fluid space for therapeutic and/or diagnostic purposes.
  • G.I gastrointestinal
  • treatment and/or diagnosis of certain diseases or disorders of the G.I. tract by means of the targeted delivery of appropriate agents, including anti-cancer or antiviral agents, is therefore facilitated.
  • Analogous methods of treatment and delivery of biologically active materials is readily contemplated for other organ and/or tissue systems.
  • the tissues are tumor or cancer tissues
  • the tetrapartate prodrugs of in the invention comprise agents suitable for treatment and/or diagnosis of such conditions.
  • the tetrapartate prodrug compositions are useful for, among other things, treating diseases which are similar to those which are treated with the parent compound(s), e.g., including compounds suitable for treating neoplastic disease, reducing tumor burden, inhibiting metastasis of tumors or neoplasms and preventing recurrences of tumor/neoplastic growths in mammals.
  • the treated animals are preferably mammals, and more preferably human patients.
  • prodrugs of the invention While veterinary use of the prodrugs of the invention will typically be employed in mammalian species, it is further contemplated that the prodrugs can also be readily employed in other species generally within the veterinary practice and animal husbandry arts, e.g., including highly valued non-mammalian exotic animals.
  • the amount of the prodrug and/or diagnostic tetrapartate tag that is administered will depend upon the amount of the parent molecule included therein. Generally, the amount of tetrapartate prodrug used in the treatment methods is that amount which effectively achieves the desired therapeutic or diagnostic result in mammals. Naturally, the dosages of the various prodrug compounds will vary somewhat depending upon the parent compound, rate of in vivo hydrolysis, molecular weight of the polymer, etc. In general, tetrapartate prodrug polymeric derivatives are administered in amounts ranging from about 5 to about 500 mg/m 2 per day, based on the native drug. The range set forth above is illustrative and those skilled in the art will determine the optimal dosing of the prodrug selected based on clinical experience and the treatment indication. Actual dosages will be apparent to the artisan without undue experimentation.
  • compositions, including prodrugs, of the present invention can be included in one or more suitable pharmaceutical compositions for administration to an animal in need thereof.
  • the pharmaceutical compositions may be in the form of a solution, suspension, tablet, capsule or the like, prepared according to methods well known in the art. It is also contemplated that administration of such compositions may be by the oral and/or parenteral routes depending upon the needs of the artisan.
  • a solution and/or suspension of the composition may be utilized, for example, as a carrier vehicle for injection or infiltration of the composition by any art known methods, e.g., by intravenous, intramuscular, subdermal injection and the like.
  • Such administration may also be by infusion into a body space or cavity, as well as by inhalation and/or intranasal routes.
  • the prodrugs are parenterally administered to animals in need thereof.
  • novel methods of treatment or administration according to the invention further includes the multi-step cleavage of the prodrug, resulting in release of the biologically active material, such as a drug or tag, within a target cell.
  • a further aspect of the invention provides the tetrapartate conjugates of the invention optionally prepared with a diagnostic tag linked to the transport enhancer described above, wherein the tag is selected for diagnostic or imaging purposes.
  • a suitable tag is prepared by linking any suitable moiety, e.g., an amino acid residue, to any art-standard emitting isotope, radio-opaque label, magnetic resonance label, or other non-radioactive isotopic labels suitable for magnetic resonance imaging, fluorescence-type labels, labels exhibiting visible colors and/or capable of fluorescing under ultraviolet, infrared or electrochemical stimulation, to allow for imaging tumor tissue during surgical procedures, and so forth.
  • the diagnostic tag is incorporated into and/or linked to a conjugated therapeutic moiety, allowing for monitoring of the distribution of a therapeutic biologically active material within an animal or human patient.
  • the inventive tagged tetrapartate conjugates are readily prepared, by art-known methods, with any suitable label, including, e.g., radioisotope labels.
  • radioisotope labels include 131 Iodine, 125 Iodine, 99m Technetium and/or 111 Indium to produce radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo.
  • radioimmunoscintigraphic agents for selective uptake into tumor cells, in vivo.
  • there are a number of art-known methods of linking peptide to Tc-99 m including, simply by way of example, those shown by U.S. Pat. Nos. 5,328,679; 5,888,474; 5,997,844; and 5,997,845, incorporated by reference herein.
  • the tetrapartate conjugate tag is administered to a patient or animal suspected of having a tumor.
  • the signal generated by the label is detected, for instance, visually, by X-ray radiography, computerized transaxial tomography, MRI, by instrumental detection of a luminescent tag, by a photo scanning device such as a gamma camera, or any other method or instrument appropriate for the nature of the selected tag.
  • the detected signal is then converted to an image or anatomical and/or physiological determination of the tumor site.
  • the image makes it possible to locate the tumor in vivo and to devise an appropriate therapeutic strategy.
  • the detected signal provides evidence of anatomical localization during treatment, providing a baseline for follow-up diagnostic and therapeutic interventions.
  • PEG employed in the following examples had a molecular weight of about 40 kDa.
  • Leucine-doxorubicin 130 mg, 0.198 mmol
  • DMAP dimethylaminopyridine
  • 121 mg, 0.99 mmol was added to a solution of compound 1, having a PEG chain of about 40 kDa (2.4 g, 0.060 mmol, FIG. 1) in 40 mL of anhydrous dimethyformamide (“DMF”).
  • DMF dimethyformamide
  • Ethyl ether ⁇ 200 mL was added to the reaction mixture to precipitate PEG derivatives, and the solid filtered and recrystallized twice from from 80 mL 2-propanol (“IPA”) to give pure product of compound 2 (1.85 g, 75%, FIG. 3 ).
  • Step 1 To a solution of compound 1 (4.5 g, 0.111 mmol, FIG. 4) in 80 mL of anhydrous methylene chloride, was added leucine t-butyl ester (248 mg, 1.11 mmol) and DMAP (136 mg, 1.11 mmol). The reaction mixture was stirred for 18 hours at room temperature. The mixture was evaporated under reduced pressure, and the residue was re-crystallized from IPA to give compound 3 (4.5 g, 99%, FIG. 4 ).
  • Step 2 compound 3 (4.70 g, 0.114 mmol) was dissolved in 22.5 mL trifluoroacetic acid and 45 mL of methylene chloride and stirred at room temperature for 2 hours. Ethyl ether was added to precipitate the PEG derivative. The crude product was filtered and washed with ethyl ether to yield compound 4 (4.4 g, 94%, FIG. 4 ).
  • Step 3 Doxorubicin (57 mg, 0.0984 mmol) and DMAP (42 mg, 0.344 mmol) were added to a solution of compound 4 (1.0 g, 0.0246 mmol, FIG. 4) in anhydrous methylene chloride (20 mL) at 0° C. for 20 minutes.
  • 1-[3-(dimethylamino) propyl]-3-ethylcarbodiimide hydrochloride (“EDC,” 28 mg, 0.148 mmol) was added, and the reaction mixture gradually warmed to room temperature and stirred overnight. The solution was filtered by gravity, and the solvent evaporated. The residual solid was recrystallized from IPA (50 mL) to give pure compound 2 (0.656 g, 67%).
  • Compound 6 (FIG. 5 ), was prepared by adding leucine-doxorubicin (130 mg, 0.198 mmol, FIG. 3) and DMAP (121 mg, 0.99 mmol) to a solution of compound 5 (2.2 g, 0.054 mmol, FIG. 5) in anhydrous DMF (30 mL). After stirring at room temperature overnight, ethyl ether ( ⁇ 100 mL) was added to the reaction mixture and the precipitated solid filtered. The solid was recrystallized twice from IPA (70 mL) to give pure product (2.09 g, 93%).
  • Compound 8 (FIG. 6 ), was prepared by the methods described in Example 2, above: by adding leucine-doxorubicin (80 mg, 0.122 mmol ) to a solution of compound 7 (1.6 g, 0.41 mmol, FIG. 4) in 30 mL of anhydrous DMF, and reacting the mixture to give the product 8, (1.44 g, 85%) which was recrystallized from IPA.
  • Compound 10 (FIG. 7A) was prepared by the methods described in Example 2, above: by adding leucine-doxorubicin (97 mg, 0.147 mmol, FIG. 7A) to a solution of compound 9 (2.0 g, 0.049 mmol, FIG. 7A) in 30 mL of anhydrous DMF. Pure compound 10 was obtained by recrystallization from IPA (1.70 g, 83%).
  • Compound 12 (FIG. 7 B), was prepared by the methods described in Example 2, above: by adding leucine-doxorubicin (134 mg, 0.204 mmol, FIG. 7B) to a solution of compound 11 (2.0 g, 0.049 mmol) ) in 30 mL of anhydrous DMF. Compound 12 was purified by recrystallization from IPA (1.69 g, 82%).
  • Step 1 To a solution of compound 1 (6.88 g, 0.170 mmol, FIG. 8 ), in 90 mL anhydrous DMF, was added 12-aminododecanoic acid (0.15 g, 0.680 mmol) and DMAP (0.114 g, 0.934 mmol). The resulting reaction mixture was stirred for 18 hours at a temperature between 50 to 60° C. The mixture was filtered, the filtrate evaporated under reduced pressure, and the residue recrystallized from IPA to give compound 13 (6.2 g, 90).
  • Step 2 To a solution of compound 13 (3.0 g, 0.074 mmol) as obtained in Step 1, above, doxorubicin hydrochloride (256 mg, 0.441 mmol), 4-methylmorpholine (“NMM”, 130 uL, 1.18 mmol), and 1-hydroxybenzotriazole hydrate (“HOBT”, 60 mg, 0.441 mmol) in 80 mL anhydrous DMF/methylene chloride (1:1) was EDC (113 mg, 0.589 mmol), and the mixture stirred at room temperature overnight. The solvent was evaporated under reduced pressure and the residue was recrystallized from IPA (100 mL) to give pure compound 14 (2.80 g, 91%)).
  • Compound 17 was prepared as illustrated by FIG. 9 .
  • Step 1 To a solution of compound 13 (5.20 g, 0.128 mmol) in 50 mL anhydrous methylene chloride was added leucine t-butyl ester (0.287 g, 1.28 mmol), DMAP (0.281 g, 2.30 mmol), and EDC (0.197 g, 1.02 mmol). The reaction mixture was stirred at room temperature for 18 hours. Solvent was removed under reduced pressure, and the residue recrystallized from IPA to yield 15 (4.8 g, 92%, FIG. 9 ). The structure was confirmed by 13 C NMR (67.8 MHz, CDCl 3 ) with peaks at ⁇ 172.22, 171.87, 155.79, 152.
  • Step 2 compound 15 (4.70 g, 0.114 mmol) was dissolved in 25 mL trifluoroacetic acid and 50 mL methylene chloride and stirred at room temperature for 2 hours. Ethyl ether was added to precipitate PEG product. The solid was filtered and washed with additional ethyl ether to yield compound 16 (4.4 g, 94%, FIG. 9 ).
  • Step 3 To a solution of compound 16 (3.3 g, 0.080 mmol), doxorubicin hydrochloride (280 mg, 0.480 mmol), NMM (130 uL, 1.18 mmol), and HOBT (74 mg, 0.480 mmol) in 90 mL of anhydrous DMF and methylene chloride (1:1) was added EDC (120 mg, 0.640 mmol), and the mixture stirred at room temperature overnight. The solvent was evaporated under reduced pressure and the residual solid was recrystallized from IPA (100 mL) to give pure compound 17 (2.90 g, 85%).
  • vehicle 20 m M sodium phosphate in 0.6% NaCl
  • Leu-Doxorubicin Leu-Doxorubicin
  • mice were sorted to evenly distribute tumor size, grouped into 6 mice/cage, and ear punched for permanent identification. Drugs were dosed intravenously via the tail vein once per week for three weeks (Qd7 ⁇ 3). Mouse weight and tumor sizes were measured at the beginning of study and twice weekly through week 5.
  • T/C percent treatment over control
  • n 6/group. ⁇ The median tumor volume of treatment and control groups were measured and compared when the control group's median tumor volume reached approximately 1000 mm 3 and one week after final dosage (day 21). T/C ⁇ 42% at 1000 mm 3 is considered significant anti-tumor activity by the Drug Evaluation Branch of the NCI.
  • TFA.Alanine-Camptothecin 18 was prepared as described in commonly assigned U.S. Pat. No. 6,127,355, Example 21, the contents of which are incorporated herein by reference.
  • Step 2 18 (185 mg, 0.36 mmol), was added to solution containing 9 (4 g, 0.0982 mmol) in 50 mL anhydrous methylene chloride and DMAP (49 mg, 0.40 mmol). The mixture was stirred at room temperature overnight. The reaction mixture was evaporated to dryness and the residual solid recrystallized two times from 2-propanol (80 mL) to give pure compound 19 (3.5 g, 86%).
  • Native bovine hemoglobin (bHb) in 100 mM sodium phosphate (pH 8.4)/65 mM NaCl buffer is modified to form the conjugated compound 23 as follows.
  • 20 mL of 22 0.8 g dissolved in 20 mM sodium phosphate/65 mM NaCl buffer at 4° C.
  • 20 mL of bHb at 4° C. 22.2 g at 11 mg/mL
  • the pH of the reaction is monitored.
  • the mixture is stirred for 1 hour, the reaction is quenched by the addition of glycine, and stirring is continued for an additional 15 minutes.
  • Cysteine (dissolved in 100 mM sodium phosphate/65 mM NaCl buffer at 4° C., 30 mM final concentration) is added to reduce oxidized hemoglobin (met-Hb) formation, and the reaction mixture is stirred for 16 hours at 4° C.
  • the PEG-Hb is diluted and diafiltered into formulation buffer (5 mM sodium bicarbonate, 4 mM Na 2 HPO 4 , 1 mM NaH2PO 4 , 150 mM NaCl, pH 7.4) to remove the unreacted PEG and/or PEG-glycine conjugate, followed by concentration to 60 mg/mL of compound 23.
  • the purity of the PEG-Hb is determined by size exclusion HPLC.

Landscapes

  • Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
  • Polyethers (AREA)
US09/758,993 1997-12-17 2001-01-12 Tetrapartate prodrugs Expired - Fee Related US6720306B2 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
US09/758,993 US6720306B2 (en) 1997-12-17 2001-01-12 Tetrapartate prodrugs
JP2002545642A JP2004518776A (ja) 2000-12-01 2001-11-30 テトラパルテートプロドラッグ
PCT/US2001/045127 WO2002043663A2 (en) 2000-12-01 2001-11-30 Tetrapartate prodrugs
CA002428018A CA2428018A1 (en) 2000-12-01 2001-11-30 Tetrapartate prodrugs
AU2002239405A AU2002239405A1 (en) 2000-12-01 2001-11-30 Tetrapartate prodrugs
EP01987164A EP1343494A4 (de) 2000-12-01 2001-11-30 Tetrapartat-prodrugs

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US99243597A 1997-12-17 1997-12-17
US09/183,557 US6180095B1 (en) 1997-12-17 1998-10-30 Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
US09/758,993 US6720306B2 (en) 1997-12-17 2001-01-12 Tetrapartate prodrugs

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/183,557 Continuation-In-Part US6180095B1 (en) 1997-12-17 1998-10-30 Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents

Publications (2)

Publication Number Publication Date
US20010031873A1 US20010031873A1 (en) 2001-10-18
US6720306B2 true US6720306B2 (en) 2004-04-13

Family

ID=26879271

Family Applications (1)

Application Number Title Priority Date Filing Date
US09/758,993 Expired - Fee Related US6720306B2 (en) 1997-12-17 2001-01-12 Tetrapartate prodrugs

Country Status (6)

Country Link
US (1) US6720306B2 (de)
EP (1) EP1037649B1 (de)
JP (1) JP4465109B2 (de)
AU (1) AU1825299A (de)
CA (1) CA2312975C (de)
WO (1) WO1999030727A1 (de)

Cited By (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040235773A1 (en) * 2003-04-13 2004-11-25 Hong Zhao Polymeric oligonucleotide prodrugs
US20060099265A1 (en) * 2003-03-20 2006-05-11 Kazuhisa Shimizu Micellar preparation containing sparingly water-soluble anticancer agent and novel block copolymer
US20060135410A1 (en) * 2003-05-29 2006-06-22 Cheng Liu Targeted delivery to legumain-expressing cells
US20080113028A1 (en) * 2004-09-22 2008-05-15 Kazuhisa Shimizu Novel Block Copolymer, Micelle Preparation, And Anticancer Agent Containing The Same As Active Ingredient
US20080159964A1 (en) * 2006-12-29 2008-07-03 Enzon Pharmaceuticals, Inc. Use of adenosine deaminase for treating pulmonary disease
US20080234183A1 (en) * 2002-06-18 2008-09-25 Mattias Hallbrink Cell Penetrating Peptides
US20080241102A1 (en) * 2004-03-23 2008-10-02 Complex Biosystems Gmbh Polymeric Prodrug with a Self-Immolative Linker
WO2008131208A1 (en) 2007-04-20 2008-10-30 Enzon Pharmaceuticals, Inc. Stable recombinant adenosine deaminase
US20090017004A1 (en) * 2007-07-11 2009-01-15 Enzon Pharmaceuticals, Inc. Polymeric drug delivery systems containing an aromatic allylic acid
US20090016985A1 (en) * 2007-07-11 2009-01-15 Enzon Pharmaceuticals, Inc. Polymeric drug delivery system containing a multi-substituted aromatic moiety
US20090047270A1 (en) * 2007-04-20 2009-02-19 Enzon Pharmaceuticals, Inc. Enzymatic anticancer therapy
US20090156629A1 (en) * 2006-06-09 2009-06-18 Enzon Pharmaceuticals, Inc. Indenoisoquinoline-releasable polymer conjugates
US20090162313A1 (en) * 2006-05-18 2009-06-25 Masayuki Kitagawa High-Molecular Weight Conjugate of Podophyllotoxins
US20090175873A1 (en) * 2005-11-29 2009-07-09 The Scripps Research Institute Inhibiting tumor cell invasion, metastasis and angiogenesis
US20090239782A1 (en) * 2006-10-03 2009-09-24 Masaharu Nakamura High-molecular weight conjugate of resorcinol derivatives
US20090281300A1 (en) * 2006-11-06 2009-11-12 Keiichiro Yamamoto High-molecular weight derivative of nucleic acid antimetabolite
US20100029849A1 (en) * 2006-11-08 2010-02-04 Keiichiro Yamamoto High molecular weight derivative of nucleic acid antimetabolite
US20100234537A1 (en) * 2006-03-28 2010-09-16 Masayuki Kitagawa Polymer conjugate of taxane
US20100279408A1 (en) * 2006-11-27 2010-11-04 Enzon Pharmaceuticals, Inc. Polymeric short interfering rna conjugates
US20100292414A1 (en) * 2007-09-28 2010-11-18 Nippon Kayaku Kabushiki Kaisha High-Molecular Weight Conjugate Of Steroids
US20110201754A1 (en) * 2008-03-18 2011-08-18 Nippon Kayaku Kabushiki Kaisha High-Molecular Weight Conjugate Of Physiologically Active Substances
DE102011118029A1 (de) 2011-06-20 2012-12-20 Universität Leipzig Modifizierte antibiotische Peptide mit variabler systemischer Freisetzung
US8808749B2 (en) 2009-05-15 2014-08-19 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of bioactive substance having hydroxy group
US9018323B2 (en) 2010-11-17 2015-04-28 Nippon Kayaku Kabushiki Kaisha Polymer derivative of cytidine metabolic antagonist
WO2015130997A1 (en) 2014-02-26 2015-09-03 Gdovin Matthew Nitrobenzaldehyde proton release for manipulation of cellular acidosis
US9149540B2 (en) 2008-05-08 2015-10-06 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of folic acid or folic acid derivative
WO2016061286A2 (en) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
US9346923B2 (en) 2011-09-11 2016-05-24 Nippon Kayaku Kabushiki Kaisha Method for manufacturing block copolymer
WO2019016240A1 (en) 2017-07-19 2019-01-24 Leadiant Biosciences Ltd ADENOSINE DEAMINASE FOR TREATING OR IMPROVING SCLERRODERMIS ASSOCIATED VASCULOPATHY
US10836774B2 (en) 2016-11-30 2020-11-17 North Carolina State University Methods for making bacteriochlorin macrocycles comprising an annulated isocyclic ring and related compounds
US10919904B2 (en) 2016-08-17 2021-02-16 North Carolina State University Northern-southern route to synthesis of bacteriochlorins

Families Citing this family (81)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6624142B2 (en) * 1997-12-30 2003-09-23 Enzon, Inc. Trimethyl lock based tetrapartate prodrugs
US7060708B2 (en) 1999-03-10 2006-06-13 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US6716452B1 (en) * 2000-08-22 2004-04-06 New River Pharmaceuticals Inc. Active agent delivery systems and methods for protecting and administering active agents
US6380405B1 (en) * 1999-09-13 2002-04-30 Nobex Corporation Taxane prodrugs
US6713454B1 (en) * 1999-09-13 2004-03-30 Nobex Corporation Prodrugs of etoposide and etoposide analogs
US6541508B2 (en) 1999-09-13 2003-04-01 Nobex Corporation Taxane prodrugs
US6413507B1 (en) 1999-12-23 2002-07-02 Shearwater Corporation Hydrolytically degradable carbamate derivatives of poly (ethylene glycol)
ES2536972T5 (es) * 2000-07-21 2022-04-06 Gilead Sciences Inc Profármacos de análogos de nucleótidos de fosfonato y métodos para seleccionar y preparar los mismos
US20020099013A1 (en) * 2000-11-14 2002-07-25 Thomas Piccariello Active agent delivery systems and methods for protecting and administering active agents
US7163918B2 (en) 2000-08-22 2007-01-16 New River Pharmaceuticals Inc. Iodothyronine compositions
US8394813B2 (en) 2000-11-14 2013-03-12 Shire Llc Active agent delivery systems and methods for protecting and administering active agents
JP2004518776A (ja) * 2000-12-01 2004-06-24 エンゾン,インコーポレーテッド テトラパルテートプロドラッグ
US7169752B2 (en) 2003-09-30 2007-01-30 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US20060014697A1 (en) 2001-08-22 2006-01-19 Travis Mickle Pharmaceutical compositions for prevention of overdose or abuse
US20070066537A1 (en) * 2002-02-22 2007-03-22 New River Pharmaceuticals Inc. Compounds and compositions for prevention of overdose of oxycodone
US7338939B2 (en) * 2003-09-30 2008-03-04 New River Pharmaceuticals Inc. Abuse-resistant hydrocodone compounds
US7375082B2 (en) * 2002-02-22 2008-05-20 Shire Llc Abuse-resistant hydrocodone compounds
EP1450844A4 (de) * 2001-11-09 2009-07-29 Enzon Inc Polymere thiol-verknüpfte prodrugs unter verwendung von benzyl-eliminationssystemen
EP1450822B1 (de) * 2001-11-09 2009-12-23 Enzon, Inc. Polyalkylenoxid-Konjugate von thiolhaltigen Arzneistoffen
WO2003051113A1 (en) 2001-12-14 2003-06-26 The University Of Wyoming Methods and compositions for controlled release of drugs
CN1176137C (zh) * 2002-01-15 2004-11-17 泛亚生物技术有限公司 多臂树杈型功能化聚乙二醇制备方法及它在药物中的应用
CN1630530A (zh) * 2002-01-18 2005-06-22 比奥根艾迪克Ma公司 聚亚烷基聚合物及其用途
AU2003211495A1 (en) * 2002-02-12 2003-09-04 Nsk Ltd. Pulley bearing for engine auxiliaries
US7700561B2 (en) * 2002-02-22 2010-04-20 Shire Llc Abuse-resistant amphetamine prodrugs
CN1650024A (zh) * 2002-02-22 2005-08-03 新河药品股份有限公司 应用肽-药物的结合减少受试者间药物血清水平的变异
US7659253B2 (en) 2002-02-22 2010-02-09 Shire Llc Abuse-resistant amphetamine prodrugs
US7105486B2 (en) * 2002-02-22 2006-09-12 New River Pharmaceuticals Inc. Abuse-resistant amphetamine compounds
CA2477004C (en) * 2002-02-22 2011-05-10 Thomas Piccariello Novel sustained release pharmaceutical compounds to prevent abuse of controlled substances
PT1534340E (pt) 2002-09-06 2012-03-13 Cerulean Pharma Inc Polímeros à base de ciclodextrina para a administração de medicamentos ligados por ligação covalente
EP1616003A4 (de) 2002-12-30 2007-06-20 Gryphon Therapeutics Inc Wasserlösliche thioester- und selenoesterverbindungen sowie verfahren zur herstellung und verwendung davon
US7332164B2 (en) 2003-03-21 2008-02-19 Enzon Pharmaceuticals, Inc. Heterobifunctional polymeric bioconjugates
SI1644019T2 (en) 2003-05-29 2018-04-30 Shire Llc AMPHETAMINE COMPOUNDS COMPOUNDS TO ABUSE
CZ294996B6 (cs) * 2003-07-16 2005-04-13 Ústav Makromolekulární Chemie Av Čr Reaktivní polymery a kopolymery na bázi N-(2-hydroxypropyl)methakrylamidu, způsob jejich přípravy a jejich použití pro syntézu polymerních léčiv, pro modifikaci biologicky aktivních proteinů a přípravu systémů pro dopravu genů
CA2534298C (en) 2003-08-01 2013-12-31 Nobex Corporation Aryl carbamate oligomers for hydrolyzable prodrugs and prodrugs comprising same
MXPA06003619A (es) * 2003-09-30 2008-01-14 New River Pharmaceuticals Inc Composiciones farmaceuticas para prevencion de sobredosis o abuso.
EP1525890A1 (de) 2003-10-02 2005-04-27 Complex Biosystems GmbH Protein-Proteophor Komplex
US20060182692A1 (en) 2003-12-16 2006-08-17 Fishburn C S Chemically modified small molecules
CA2549730C (en) * 2003-12-16 2012-02-14 Nektar Therapeutics Al, Corporation Chemically modified small molecules
US7968085B2 (en) 2004-07-05 2011-06-28 Ascendis Pharma A/S Hydrogel formulations
WO2006105993A2 (en) 2005-04-05 2006-10-12 Istituto Di Ricerche Di Biologia Molecolare P Angeletti Spa Method for shielding functional sites or epitopes on proteins
GB2427360A (en) 2005-06-22 2006-12-27 Complex Biosystems Gmbh Aliphatic prodrug linker
KR20080091180A (ko) * 2005-12-30 2008-10-09 파마이센시아 코퍼레이션 약물-폴리머 컨쥬게이트
JP2010503707A (ja) * 2006-09-15 2010-02-04 エンゾン ファーマスーティカルズ インコーポレイテッド オリゴヌクレオチドの送達を目的としたヒンダードエステル系生体分解性リンカー
CN101534861B (zh) 2006-09-15 2013-10-02 安佐制药股份有限公司 具有基于位阻酯的生物可降解连接基的聚环氧烷
EP2076257A4 (de) * 2006-09-15 2014-04-16 Belrose Pharma Inc Polymere konjugate mit positiv geladenen teilen
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
TW201509433A (zh) * 2006-12-13 2015-03-16 Cerulean Pharma Inc 用於醫物傳遞之以環糊精為基之聚合物
JP2010516625A (ja) * 2007-01-24 2010-05-20 インサート セラピューティクス, インコーポレイテッド 制御された薬物送達のためのテザー基を有するポリマー−薬物コンジュゲート
CZ300776B6 (cs) * 2007-11-21 2009-08-05 Univerzita Pardubice Polymerní konjugát antimykotického léciva, zpusob jeho prípravy a farmaceutická kompozice jej obsahující
WO2009114811A2 (en) * 2008-03-14 2009-09-17 Merit Medical Systems, Inc. Securement apparatus
PT3050576T (pt) 2008-04-29 2021-06-01 Ascendis Pharma Growth Disorders Div A/S Compostos recombinantes e peguilados da hormona de crescimento humana
EP2113256A1 (de) 2008-04-29 2009-11-04 Ascendis Pharma AS Pegylierte rhGH-Verbindungen
US20120058084A1 (en) 2009-03-05 2012-03-08 Ascendis Pharma As Interferon Alpha Carrier Prodrugs
KR20120089843A (ko) 2009-07-31 2012-08-14 사노피-아벤티스 도이칠란트 게엠베하 인슐린 링커 접합체를 포함한 프로드럭
EP2459227B1 (de) * 2009-07-31 2021-03-17 Ascendis Pharma A/S Prodrugs mit einem aromatischen Amin, die durch eine Amidbindung mit einer Trägersubstanz verbunden sind
DK2459171T3 (en) 2009-07-31 2017-09-25 Sanofi Aventis Deutschland Long-acting insulin composition
EP2470190A4 (de) * 2009-08-27 2013-07-17 Seachaid Pharmaceuticals Inc Echinocandinderivate
DK2512450T3 (en) 2009-12-15 2018-04-23 Ascendis Pharma Endocrinology Div A/S Dry growth hormone composition transiently bound to a polymer carrier
US9062094B2 (en) 2010-01-22 2015-06-23 Ascendis Pharma As Dipeptide-based prodrug linkers for aliphatic amine-containing drugs
US20130030359A1 (en) 2010-01-22 2013-01-31 Ascendis Pharma A/S Dipeptide-based prodrug linkers for aromatic amine-containing drugs
EP2438930A1 (de) 2010-09-17 2012-04-11 Sanofi-Aventis Deutschland GmbH Prodrug mit einem exendinbindenden Konjugat
EA023929B1 (ru) 2010-09-30 2016-07-29 Астразенека Аб Кристаллический конъюгат налоксол-peg
PT2717917T (pt) 2011-07-05 2016-07-27 Bioasis Technologies Inc Conjugados de anticorpos p97
CA3140358A1 (en) 2012-07-31 2014-02-06 Bioasis Technologies, Inc. Dephosphorylated lysosomal storage disease proteins and methods of use thereof
WO2014055493A1 (en) 2012-10-02 2014-04-10 Cerulean Pharma Inc. Methods and systems for polymer precipitation and generation of particles
CN104884056A (zh) 2012-11-12 2015-09-02 雷德伍德生物科技股份有限公司 用于制备缀合物的化合物和方法
US9310374B2 (en) 2012-11-16 2016-04-12 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
AU2013344464A1 (en) 2012-11-16 2015-05-21 The Regents Of The University Of California Pictet-Spengler ligation for protein chemical modification
AU2014243816B2 (en) 2013-03-13 2019-01-31 Bioasis Technologies Inc. Fragments of p97 and uses thereof
US20150093399A1 (en) 2013-08-28 2015-04-02 Bioasis Technologies, Inc. Cns-targeted conjugates having modified fc regions and methods of use thereof
EP3074010A4 (de) 2013-11-27 2017-10-25 Redwood Bioscience, Inc. Hydrazinylpyrrolverbindungen und verfahren zur herstellung eines konjugats
EP3604384B1 (de) 2017-03-30 2021-09-08 NOF Corporation Hydrophiles polymerderivat mit selbstimmolativem acetallinker und verbundstoff damit
CN109589415B (zh) * 2017-09-30 2022-02-18 天津键凯科技有限公司 一种聚乙二醇-多肽和蛋白类药物的结合物
CN111094462A (zh) 2017-12-26 2020-05-01 贝克顿·迪金森公司 深紫外线可激发的水溶剂化聚合物染料
EP3775052B1 (de) 2018-03-30 2024-06-05 Becton, Dickinson and Company Wasserlösliche polymere farbstoffe mit seitenständigen chromophoren
WO2020023300A1 (en) 2018-07-22 2020-01-30 Bioasis Technologies, Inc. Treatment of lymmphatic metastases
US20210355468A1 (en) 2020-05-18 2021-11-18 Bioasis Technologies, Inc. Compositions and methods for treating lewy body dementia
US20210393787A1 (en) 2020-06-17 2021-12-23 Bioasis Technologies, Inc. Compositions and methods for treating frontotemporal dementia
EP4155349A1 (de) 2021-09-24 2023-03-29 Becton, Dickinson and Company Wasserlösliche gelbe grünabsorbierende farbstoffe
WO2024007016A2 (en) 2022-07-01 2024-01-04 Beckman Coulter, Inc. Novel fluorescent dyes and polymers from dihydrophenanthrene derivatives
WO2024044327A1 (en) 2022-08-26 2024-02-29 Beckman Coulter, Inc. Dhnt monomers and polymer dyes with modified photophysical properties

Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US5093531A (en) 1988-05-06 1992-03-03 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol substituted phenyl glyoxals
US5122614A (en) 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5321095A (en) 1993-02-02 1994-06-14 Enzon, Inc. Azlactone activated polyalkylene oxides
US5349001A (en) 1993-01-19 1994-09-20 Enzon, Inc. Cyclic imide thione activated polyalkylene oxides
US5382657A (en) 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
WO1996022277A1 (en) 1995-01-19 1996-07-25 Cancer Research Campaign Technology Limited Nitrogen mustard prodrugs with novel lipophilic protecting groups, and processes for their production
US5561119A (en) 1991-04-30 1996-10-01 Laboratoires Hoechst Glycosylated prodrugs, their method of preparation and their uses
US5569720A (en) 1992-06-19 1996-10-29 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5605976A (en) 1995-05-15 1997-02-25 Enzon, Inc. Method of preparing polyalkylene oxide carboxylic acids
US5614549A (en) 1992-08-21 1997-03-25 Enzon, Inc. High molecular weight polymer-based prodrugs
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5672584A (en) 1995-04-25 1997-09-30 The University Of Kansas Cyclic prodrugs of peptides and peptide nucleic acids having improved metabolic stability and cell membrane permeability
US5710135A (en) 1995-06-27 1998-01-20 Pharmachemie B.V. Anthracycline prodrugs, method for preparation as well as their use in selective chemotherapy
WO1998013059A1 (en) 1996-09-27 1998-04-02 Bristol-Myers Squibb Company Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
US5840900A (en) 1993-10-20 1998-11-24 Enzon, Inc. High molecular weight polymer-based prodrugs
US5877158A (en) * 1993-09-22 1999-03-02 Behringwerke Aktiengesellschaft Pro-prodrugs, their production and use
US5965119A (en) 1997-12-30 1999-10-12 Enzon, Inc. Trialkyl-lock-facilitated polymeric prodrugs of amino-containing bioactive agents
US6180095B1 (en) * 1997-12-17 2001-01-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
WO2001009139A1 (en) 1999-08-03 2001-02-08 The Stehlin Foundation For Cancer Research Aromatic esters of camptothecins and methods to treat cancers

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP3026133B2 (ja) * 1992-11-05 2000-03-27 コニカ株式会社 ハロゲン化銀写真感光材料

Patent Citations (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
US5093531A (en) 1988-05-06 1992-03-03 Sumitomo Pharmaceuticals Company, Limited Polyethylene glycol substituted phenyl glyoxals
US5122614A (en) 1989-04-19 1992-06-16 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
US5561119A (en) 1991-04-30 1996-10-01 Laboratoires Hoechst Glycosylated prodrugs, their method of preparation and their uses
US5569720A (en) 1992-06-19 1996-10-29 Farmitalia Carlo Erba S.R.L. Polymer-bound paclitaxel derivatives
US5614549A (en) 1992-08-21 1997-03-25 Enzon, Inc. High molecular weight polymer-based prodrugs
US5382657A (en) 1992-08-26 1995-01-17 Hoffmann-La Roche Inc. Peg-interferon conjugates
US5349001A (en) 1993-01-19 1994-09-20 Enzon, Inc. Cyclic imide thione activated polyalkylene oxides
US5321095A (en) 1993-02-02 1994-06-14 Enzon, Inc. Azlactone activated polyalkylene oxides
US5877158A (en) * 1993-09-22 1999-03-02 Behringwerke Aktiengesellschaft Pro-prodrugs, their production and use
US5840900A (en) 1993-10-20 1998-11-24 Enzon, Inc. High molecular weight polymer-based prodrugs
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
WO1996022277A1 (en) 1995-01-19 1996-07-25 Cancer Research Campaign Technology Limited Nitrogen mustard prodrugs with novel lipophilic protecting groups, and processes for their production
US5672584A (en) 1995-04-25 1997-09-30 The University Of Kansas Cyclic prodrugs of peptides and peptide nucleic acids having improved metabolic stability and cell membrane permeability
US5605976A (en) 1995-05-15 1997-02-25 Enzon, Inc. Method of preparing polyalkylene oxide carboxylic acids
US5710135A (en) 1995-06-27 1998-01-20 Pharmachemie B.V. Anthracycline prodrugs, method for preparation as well as their use in selective chemotherapy
WO1998013059A1 (en) 1996-09-27 1998-04-02 Bristol-Myers Squibb Company Hydrolyzable prodrugs for delivery of anticancer drugs to metastatic cells
US6180095B1 (en) * 1997-12-17 2001-01-30 Enzon, Inc. Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
US5965119A (en) 1997-12-30 1999-10-12 Enzon, Inc. Trialkyl-lock-facilitated polymeric prodrugs of amino-containing bioactive agents
WO2001009139A1 (en) 1999-08-03 2001-02-08 The Stehlin Foundation For Cancer Research Aromatic esters of camptothecins and methods to treat cancers

Non-Patent Citations (15)

* Cited by examiner, † Cited by third party
Title
Bundgaard, H. The Double Prodrug Concept and its Applications, Advanced Drug Delivery Reviews, 3 1989 pp. 39-65.
Carl, P.L., et al. A Novel Connector Linkage Applicable in Prodrug Design, Journal of Medicinal Chemistry, vol. 24, No. 5 (May 1981).
Declaration under 37 C.F.R. 1.132 of Jeffrey McGuire, Sep. 8, 1999.
Declaration under 37 C.F.R. 1.132 of Richard B. Greenwald including Exhibits 1 and 2, Sep. 8, 1999.
Greenwald, Richard B., et al. Drug Delivery Systems Employing 1,4-or 1,6-Elimination: Poly (ethylene-glycol) Prodrugs of Amine-Containing Compounds, Journal of Medicinal Chemistry, vol. 42, No. 18, pp. 3657-3667, 1999.
Jungheim, L.N. et al., Design of Antitumor Prodrugs: Substrates for Antibody Targeted Enzymes, Chem. Rev. 1994; 94 pp. 1553-1566.
Leenders, R.G.G. et al. B-Glucuronyl Carbamate Based Pro-moieties Designed for Prodrugs in ADEPT, 1995; Tetrahedron Letters vol. 36, No. 10 pp. 1701-1704.
Leenders, R.G.G.et al., Highly Diastereoselective Synthesis of Anomeric B-O-Glycopyranosyl Carbamates from Isocyanates, Synthesis Nov. 1996; pp 1309-1312.
Maeda, H., et al., "Tumor vascular permeability and the EPR effect in macromolecular therapeutics: a review" J. Contr. Release 65 (2000) pp. 271-284.
Record of Invention "2- and 4- Hydroxybenzyl Alcohol-Containing Linkers", Zalipsky, Nov. 18, 1991.
Shan, D., et al., Prodrug Strategies Based on Intramolecular Cyclization Reactions, Journal of Pharmaceutical Sciences, Jul. 1997, vol. 86, No. 7, pp765-767.
Shearwater Polymers, Inc., Catalog-Polyethylene Glycol Derivatives, 1997-1998; p. 8.
Wakselman, M. et al., An Alkali-labile Substituted Benzyloxycarbonyl Amino-protecting Group, JCS Chem. Comm1973; pp. 593-594.
Waldmann, H. et al, Synthesis of the Palmitoylated and Farnesylated C-Terminal Lipohexapeptide of the Human N-Ras Protein by Employing . . . , Angew. Chem Int. Ed. 1995, 34 No. 20 ; pp. 2259-2262.
Zier et al. Polyethylene Glycol Bound Benzyl and Fluorenyl Derivatives . . . Tetrahedron Letters, vol. 35, No. 7, pp. 1039-1042 (1994).

Cited By (54)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080234183A1 (en) * 2002-06-18 2008-09-25 Mattias Hallbrink Cell Penetrating Peptides
US20060099265A1 (en) * 2003-03-20 2006-05-11 Kazuhisa Shimizu Micellar preparation containing sparingly water-soluble anticancer agent and novel block copolymer
US20040235773A1 (en) * 2003-04-13 2004-11-25 Hong Zhao Polymeric oligonucleotide prodrugs
US20090318538A1 (en) * 2003-04-13 2009-12-24 Enzon Pharmaceuticals, Inc. Polymeric oligonucleotide prodrugs
US8168605B2 (en) 2003-04-13 2012-05-01 Enzon Pharmaceuticals, Inc. Polymeric oligonucleotide prodrugs
US7595304B2 (en) 2003-04-13 2009-09-29 Enzon Pharmaceuticals, Inc. Polymeric oligonucleotide prodrugs
US20060135410A1 (en) * 2003-05-29 2006-06-22 Cheng Liu Targeted delivery to legumain-expressing cells
US7608591B2 (en) 2003-05-29 2009-10-27 The Scripps Research Institute Targeted delivery to legumain-expressing cells
US20080241102A1 (en) * 2004-03-23 2008-10-02 Complex Biosystems Gmbh Polymeric Prodrug with a Self-Immolative Linker
US8377917B2 (en) 2004-03-23 2013-02-19 Complex Biosystems Gmbh Polymeric prodrug with a self-immolative linker
US9238686B2 (en) 2004-03-23 2016-01-19 Ascendis Pharma Gmbh Polymeric prodrug with self-immolative linker
US20080113028A1 (en) * 2004-09-22 2008-05-15 Kazuhisa Shimizu Novel Block Copolymer, Micelle Preparation, And Anticancer Agent Containing The Same As Active Ingredient
US9434822B2 (en) 2004-09-22 2016-09-06 Nippon Kayaku Kabushiki Kaisha Block copolymer, micelle preparation, and anticancer agent containing the same as active ingredient
US20090175873A1 (en) * 2005-11-29 2009-07-09 The Scripps Research Institute Inhibiting tumor cell invasion, metastasis and angiogenesis
US20100234537A1 (en) * 2006-03-28 2010-09-16 Masayuki Kitagawa Polymer conjugate of taxane
US8323669B2 (en) 2006-03-28 2012-12-04 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of taxane
US8940332B2 (en) 2006-05-18 2015-01-27 Nippon Kayaku Kabushiki Kaisha High-molecular weight conjugate of podophyllotoxins
US20090162313A1 (en) * 2006-05-18 2009-06-25 Masayuki Kitagawa High-Molecular Weight Conjugate of Podophyllotoxins
US20090156629A1 (en) * 2006-06-09 2009-06-18 Enzon Pharmaceuticals, Inc. Indenoisoquinoline-releasable polymer conjugates
US20090239782A1 (en) * 2006-10-03 2009-09-24 Masaharu Nakamura High-molecular weight conjugate of resorcinol derivatives
US20090281300A1 (en) * 2006-11-06 2009-11-12 Keiichiro Yamamoto High-molecular weight derivative of nucleic acid antimetabolite
US8334364B2 (en) 2006-11-06 2012-12-18 Nipon Kayaku Kabushiki Kaisha High-molecular weight derivative of nucleic acid antimetabolite
US8188222B2 (en) 2006-11-08 2012-05-29 Nippon Kayaku Kabushiki Kaisha High molecular weight derivative of nucleic acid antimetabolite
US20100029849A1 (en) * 2006-11-08 2010-02-04 Keiichiro Yamamoto High molecular weight derivative of nucleic acid antimetabolite
US20100279408A1 (en) * 2006-11-27 2010-11-04 Enzon Pharmaceuticals, Inc. Polymeric short interfering rna conjugates
US20080159964A1 (en) * 2006-12-29 2008-07-03 Enzon Pharmaceuticals, Inc. Use of adenosine deaminase for treating pulmonary disease
US8741283B2 (en) 2007-04-20 2014-06-03 Sigma-Tau Rare Diseases, S.A. Adenosine deaminase anticancer therapy
US8071741B2 (en) 2007-04-20 2011-12-06 Defiante Farmaceutica, S.A. Stable recombinant adenosine deaminase
US20090047271A1 (en) * 2007-04-20 2009-02-19 Enzon Pharmaceuticals, Inc. Stable recombinant adenosine deaminase
US20090047270A1 (en) * 2007-04-20 2009-02-19 Enzon Pharmaceuticals, Inc. Enzymatic anticancer therapy
WO2008131208A1 (en) 2007-04-20 2008-10-30 Enzon Pharmaceuticals, Inc. Stable recombinant adenosine deaminase
US20090017004A1 (en) * 2007-07-11 2009-01-15 Enzon Pharmaceuticals, Inc. Polymeric drug delivery systems containing an aromatic allylic acid
US20090016985A1 (en) * 2007-07-11 2009-01-15 Enzon Pharmaceuticals, Inc. Polymeric drug delivery system containing a multi-substituted aromatic moiety
USRE46190E1 (en) 2007-09-28 2016-11-01 Nippon Kayaku Kabushiki Kaisha High-molecular weight conjugate of steroids
US8703878B2 (en) 2007-09-28 2014-04-22 Nippon Kayaku Kabushiki Kaisha High-molecular weight conjugate of steroids
US20100292414A1 (en) * 2007-09-28 2010-11-18 Nippon Kayaku Kabushiki Kaisha High-Molecular Weight Conjugate Of Steroids
US8920788B2 (en) 2008-03-18 2014-12-30 Nippon Kayaku Kabushiki Kaisha High-molecular weight conjugate of physiologically active substances
US20110201754A1 (en) * 2008-03-18 2011-08-18 Nippon Kayaku Kabushiki Kaisha High-Molecular Weight Conjugate Of Physiologically Active Substances
US9149540B2 (en) 2008-05-08 2015-10-06 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of folic acid or folic acid derivative
US8808749B2 (en) 2009-05-15 2014-08-19 Nippon Kayaku Kabushiki Kaisha Polymer conjugate of bioactive substance having hydroxy group
US9018323B2 (en) 2010-11-17 2015-04-28 Nippon Kayaku Kabushiki Kaisha Polymer derivative of cytidine metabolic antagonist
DE102011118029A1 (de) 2011-06-20 2012-12-20 Universität Leipzig Modifizierte antibiotische Peptide mit variabler systemischer Freisetzung
US9387255B2 (en) 2011-06-20 2016-07-12 Universitaet Leipzig Modified antibiotic peptides having variable systemic release
WO2012175532A1 (de) 2011-06-20 2012-12-27 Universität Leipzig Modifizierte antibiotische peptide mit variabler systemischer freisetzung
US9856298B2 (en) 2011-06-20 2018-01-02 Universitaet Leipzig Modified antibiotic peptides having variable systemic release
US9346923B2 (en) 2011-09-11 2016-05-24 Nippon Kayaku Kabushiki Kaisha Method for manufacturing block copolymer
WO2015130997A1 (en) 2014-02-26 2015-09-03 Gdovin Matthew Nitrobenzaldehyde proton release for manipulation of cellular acidosis
US10946096B2 (en) 2014-02-26 2021-03-16 The Board Of Regents Of The University Of Texas System Nitrobenzaldehyde proton release for manipulation of cellular acidosis
WO2016061286A2 (en) 2014-10-14 2016-04-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
US9969998B2 (en) 2014-10-14 2018-05-15 Halozyme, Inc. Compositions of adenosine deaminase-2 (ADA2), variants thereof and methods of using same
US11584923B2 (en) 2014-10-14 2023-02-21 Halozyme, Inc. Compositions of adenosine deaminase-2 (ADA2), variants thereof and methods of using same
US10919904B2 (en) 2016-08-17 2021-02-16 North Carolina State University Northern-southern route to synthesis of bacteriochlorins
US10836774B2 (en) 2016-11-30 2020-11-17 North Carolina State University Methods for making bacteriochlorin macrocycles comprising an annulated isocyclic ring and related compounds
WO2019016240A1 (en) 2017-07-19 2019-01-24 Leadiant Biosciences Ltd ADENOSINE DEAMINASE FOR TREATING OR IMPROVING SCLERRODERMIS ASSOCIATED VASCULOPATHY

Also Published As

Publication number Publication date
CA2312975A1 (en) 1999-06-24
EP1037649A1 (de) 2000-09-27
EP1037649A4 (de) 2004-11-17
EP1037649B1 (de) 2009-09-30
JP4465109B2 (ja) 2010-05-19
WO1999030727A9 (en) 1999-09-23
CA2312975C (en) 2012-08-21
WO1999030727A1 (en) 1999-06-24
AU1825299A (en) 1999-07-05
JP2002508400A (ja) 2002-03-19
US20010031873A1 (en) 2001-10-18

Similar Documents

Publication Publication Date Title
US6720306B2 (en) Tetrapartate prodrugs
US6624142B2 (en) Trimethyl lock based tetrapartate prodrugs
US6180095B1 (en) Polymeric prodrugs of amino- and hydroxyl-containing bioactive agents
US7122189B2 (en) Releasable polymeric conjugates based on aliphatic biodegradable linkers
US7087229B2 (en) Releasable polymeric conjugates based on aliphatic biodegradable linkers
US6303569B1 (en) Trialkyl-lock-facilitated polymeric prodrugs of amino-containing bioactive agents
US8192743B2 (en) Releasable polymeric conjugates based on aliphatic biodegradable linkers
US20090203706A1 (en) Lysine-based polymeric linkers
CA2465090C (en) Polymeric thiol-linked prodrugs employing benzyl elimination systems
CA2437986A1 (en) Terminally-branched polymeric linkers and polymeric conjugates containing the same
US6936597B2 (en) Prodrugs of anticancer agents employing substituted aromatic acids
EP1343494A2 (de) Tetrapartat-prodrugs
WO2017199046A1 (en) Conjugates and conjugating reagents

Legal Events

Date Code Title Description
AS Assignment

Owner name: ENZON, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:GREENWALD, RICHARD B.;ZHAO, HONG;REEL/FRAME:011738/0418

Effective date: 20010411

CC Certificate of correction
FPAY Fee payment

Year of fee payment: 4

FPAY Fee payment

Year of fee payment: 8

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20160413